1
|
Behzadi P, Chandran D, Chakraborty C, Bhattacharya M, Saikumar G, Dhama K, Chakraborty A, Mukherjee S, Sarshar M. The dual role of toll-like receptors in COVID-19: Balancing protective immunity and immunopathogenesis. Int J Biol Macromol 2025; 284:137836. [PMID: 39613064 DOI: 10.1016/j.ijbiomac.2024.137836] [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: 06/09/2024] [Revised: 11/01/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Toll-like receptors (TLRs) of human are considered as the most critical immunological mediators of inflammatory pathogenesis of COVID-19. These immunoregulatory glycoproteins are located on the surface and/or intracellular compartment act as innate immune sensors. Upon binding with distinct SARS-CoV-2 ligand(s), TLRs signal activation of different transcription factors that induce expression of the proinflammatory mediators that collectively induce 'cytokine storm'. Similarly, TLR activation is also pivotal in conferring protection to infection and invasion as well as upregulating the tissue repair pathways. This dual role of the human TLRs in deciding the fate of SARS-CoV-2 has made these receptor proteins as the critical mediators of immunoprotective and immunopathogenic consequences associated with COVID-19. Herein, pathbreaking discoveries exploring the immunobiological importance of the TLRs in COVID-19 and developing TLR-directed therapeutic intervention have been reviewed by accessing the up-to-date literatures available in the public domain/databases. In accordance with our knowledge in association with the importance of TLRs' role against viruses and identification of viral particles, they have been recognized as suitable candidates with high potential as vaccine adjuvants. In this regard, the agonists of TLR4 and TLR9 have effective potential in vaccine technology while the others need further investigations. This comprehensive review suggests that basal level expression of TLRs can act as friends to keep our body safe from strangers but act as a foe via overexpression. Therefore, selective inhibition of the overexpressed TLRs appears to be a solution to counteract the cytokine storm while TLR-agonists as vaccine adjuvants could lessen the risk of infection in the naïve population.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 37541-374, Iran.
| | | | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, VyasaVihar, Balasore, 756020, Odisha, India
| | - Guttula Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| | - Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India.
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, 00146, Rome, Italy
| |
Collapse
|
2
|
Bogacheva MS, Kuivanen S, Potdar S, Hassinen A, Huuskonen S, Pöhner I, Luck TJ, Turunen L, Feodoroff M, Szirovicza L, Savijoki K, Saarela J, Tammela P, Paavolainen L, Poso A, Varjosalo M, Kallioniemi O, Pietiäinen V, Vapalahti O. Drug repurposing platform for deciphering the druggable SARS-CoV-2 interactome. Antiviral Res 2024; 223:105813. [PMID: 38272320 DOI: 10.1016/j.antiviral.2024.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has heavily challenged the global healthcare system. Despite the vaccination programs, the new virus variants are circulating. Further research is required for understanding of the biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and for discovery of therapeutic agents against the virus. Here, we took advantage of drug repurposing to identify if existing drugs could inhibit SARS-CoV-2 infection. We established an open high throughput platform for in vitro screening of drugs against SARS-CoV-2 infection. We screened ∼1000 drugs for their ability to inhibit SARS-CoV-2-induced cell death in the African green monkey kidney cell line (Vero-E6), analyzed how the hit compounds affect the viral N (nucleocapsid) protein expression in human cell lines using high-content microscopic imaging and analysis, determined the hit drug targets in silico, and assessed their ability to cause phospholipidosis, which can interfere with the viral replication. Duvelisib was found by in silico interaction assay as a potential drug targeting virus-host protein interactions. The predicted interaction between PARP1 and S protein, affected by Duvelisib, was further validated by immunoprecipitation. Our results represent a rapidly applicable platform for drug repurposing and evaluation of the new emerging viruses' responses to the drugs. Further in silico studies help us to discover the druggable host pathways involved in the infectious cycle of SARS-CoV-2.
Collapse
Affiliation(s)
- Mariia S Bogacheva
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland.
| | - Suvi Kuivanen
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Hassinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sini Huuskonen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ina Pöhner
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tamara J Luck
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Michaela Feodoroff
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Leonora Szirovicza
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Kirsi Savijoki
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Päivi Tammela
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Poso
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Department of Internal Medicine VIII, University Hospital Tubingen, Tubingen, Germany
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland; Science for Life Laboratory (SciLifeLab), Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland; HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
3
|
Asghari F, Asghary A, Majidi Zolbanin N, Faraji F, Jafari R. Immunosenescence and Inflammaging in COVID-19. Viral Immunol 2023; 36:579-592. [PMID: 37797216 DOI: 10.1089/vim.2023.0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Abstract
Despite knowledge gaps in understanding the full spectrum of the hyperinflammatory phase caused by SARS-CoV-2, according to the World Health Organization (WHO), COVID-19 is still the leading cause of death worldwide. Susceptible people to severe COVID-19 are those with underlying medical conditions or those with dysregulated and senescence-associated immune responses. As the immune system undergoes aging in the elderly, such drastic changes predispose them to various diseases and affect their responsiveness to infections, as seen in COVID-19. At-risk groups experience poor prognosis in terms of disease recovery. Changes in the quantity and quality of immune cell function have been described in numerous literature sites. Impaired immune cell function along with age-related metabolic changes can lead to features such as hyperinflammatory response, immunosenescence, and inflammaging in COVID-19. Inflammaging is related to the increased activity of the most inflammatory factors and is the main cause of age-related diseases and tissue failure in the elderly. Since hyperinflammation is a common feature of most severe cases of COVID-19, this pathway, which is not fully understood, leads to immunosenescence and inflammaging in some individuals, especially in the elderly and those with comorbidities. In this review, we shed some light on the age-related abnormalities of innate and adaptive immune cells and how hyperinflammatory immune responses contribute to the inflammaging process, leading to clinical deterioration. Further, we provide insights into immunomodulation-based therapeutic approaches, which are potentially important considerations in vaccine design for elderly populations.
Collapse
Affiliation(s)
- Faezeh Asghari
- Department of Immunology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amir Asghary
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
Petakh P, Kamyshna I, Kamyshnyi A. Unveiling the potential pleiotropic effects of metformin in treating COVID-19: a comprehensive review. Front Mol Biosci 2023; 10:1260633. [PMID: 37881440 PMCID: PMC10595158 DOI: 10.3389/fmolb.2023.1260633] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023] Open
Abstract
This review article explores the potential of metformin, a medication commonly used for type 2 diabetes, as an antiviral and anti-inflammatory agent in the context of coronavirus disease 2019 (COVID-19). Metformin has demonstrated inhibitory effects on the growth of SARS-CoV-2 in cell culture models and has shown promising results in reducing viral load and achieving undetectable viral levels in clinical trials. Additionally, metformin exhibits anti-inflammatory properties by reducing the production of pro-inflammatory cytokines and modulating immune cell function, which may help prevent cytokine storms associated with severe COVID-19. The drug's ability to regulate the balance between pro-inflammatory Th17 cells and anti-inflammatory Treg cells suggests its potential in mitigating inflammation and restoring T cell functionality. Furthermore, metformin's modulation of the gut microbiota, particularly changes in bacterial taxa and the production of short-chain fatty acids, may contribute to its therapeutic effects. The interplay between metformin, bile acids, the gut microbiome, glucagon-like peptide-1 secretion, and glycemic control has implications for the management of diabetes and potential interventions in COVID-19. By refreshing the current evidence, this review highlights the potential of metformin as a therapeutic option in the management of COVID-19, while also exploring its effects on the gut microbiome and immunometabolism.
Collapse
Affiliation(s)
- Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Aleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
5
|
Tran NT, Liang H, Li J, Deng T, Bakky MAH, Zhang M, Li S. Cellular responses in crustaceans under white spot syndrome virus infection. FISH & SHELLFISH IMMUNOLOGY 2023; 140:108984. [PMID: 37549875 DOI: 10.1016/j.fsi.2023.108984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Innate immunity plays the most important system responsible for protecting crustaceans against invading pathogens. White spot syndrome virus (WSSV) is considered a serious pathogen in crustaceans with high cumulative mortality and morbidity in infected animals. Understanding the mechanism of the response of hosts to WSSV infection is necessary, which is useful for effective prevention in controlling infection. In this review, we summarize the participation of signaling pathways (toll, immune deficiency, JAK/STAT, endocytosis, mitogen-activated protein kinase, PI3K/Akt/mTOR, cGAS-STING, Wingless/Integrated signal transduction, and prophenoloxidase (proPO) cascade) and the activity of cells (apoptosis, autophagy, as well as, reactive oxygen species and antioxidant enzymes) in the cellular-mediated immune response of crustaceans during WSSV infection. The information presented in this current review is important for a better understanding of the mechanism of the response of hosts to pathogens. Additionally, this provides a piece of basic knowledge for discovering approaches to strengthen the immune system and resistance of cultured animals against viral infections.
Collapse
Affiliation(s)
- Ngoc Tuan Tran
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China.
| | - Huifen Liang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Jinkun Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Taoqiu Deng
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Md Akibul Hasan Bakky
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Ming Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China.
| |
Collapse
|
6
|
Shi Y, Wang M, Wu L, Li X, Liao Z. COVID-19 associated liver injury: An updated review on the mechanisms and management of risk groups. LIVER RESEARCH 2023; 7:207-215. [PMID: 39958382 PMCID: PMC11792068 DOI: 10.1016/j.livres.2023.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/11/2023] [Accepted: 07/09/2023] [Indexed: 09/02/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has been associated with various liver injury cases worldwide. To date, the prevalence, mechanism, clinical manifestations, diagnosis, and outcomes of COVID-19-induced liver injury in various at-risk groups are not well defined. Liver injury may arise in the prevention and treatment of COVID-19 from direct causes such as viral infection and indirect causes such as systemic inflammation, hypoxic changes, and drugs that exacerbate any pre-existing liver disease. Studies have found that patients with underlying liver disease are at higher risk of COVID-19-induced liver injury. Certain condition of cardiopulmonary and metabolic diseases and vulnerable stages in lifespan may also involve in the development of COVID-19-induced liver injury. This review summarized studies of COVID-19-induced liver injury in different at-risk groups regarding their clinical characteristics, parameters, and correlations of the severity with these indicators and signs as well as potential treatment suggestions, to increase attention to physiological and pathological conditions and continue liver function monitoring as they can help in strengthening early supportive treatment and reducing the incidence of adverse outcomes.
Collapse
Affiliation(s)
- Yue Shi
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Liqun Wu
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Xuexin Li
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Lee CYS, Suzuki JB. COVID-19: Variants, Immunity, and Therapeutics for Non-Hospitalized Patients. Biomedicines 2023; 11:2055. [PMID: 37509694 PMCID: PMC10377623 DOI: 10.3390/biomedicines11072055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The continuing transmission of coronavirus disease 2019 (COVID-19) remains a world-wide 21st-century public health emergency of concern. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused greater than 600 million cases of COVID-19 and over 6 million deaths globally. COVID-19 continues to be a highly transmissible disease despite efforts by public health officials and healthcare providers to manage and control the disease. Variants identified in selected worldwide epicenters add to the complexity of vaccine efficacy, overage, and antibody titer maintenance and bioactivity. The identification of the SARS-CoV-2 variants is described with respect to evading protective efficacy of COVID-19 vaccines and breakthrough infections. Vaccines and other therapeutics have prevented millions of SARS-CoV-2 infections and thousands of deaths in the United States. We explore aspects of the immune response in a condensed discussion to understand B and T cell lymphocyte regulatory mechanisms and antibody effectiveness and senescence. Finally, COVID-19 therapies including Paxlovid, Remdisivir, Molnupiravir and convalescent plasma in non-hospitalized patients are presented with limitations for identification, collection, and distribution to infected patients.
Collapse
Affiliation(s)
- Cameron Y S Lee
- Private Practice in Oral, Maxillofacial and Reconstructive Surgery, Aiea, HI 96701, USA
- Department of Periodontology and Oral Implantology, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA
| | - Jon B Suzuki
- Department of Periodontology and Oral Implantology, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA
- Department of Graduate Periodontics, University of Maryland, Baltimore, MD 20742, USA
- Department of Graduate Prosthodontics, University of Washington, Seattle, WA 98195, USA
- Department of Graduate Periodontics, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
8
|
Li J, Han J, Shi Y, Liu M. Rapamycin inhibits corneal inflammatory response and neovascularization in a mouse model of corneal alkali burn. Exp Eye Res 2023:109539. [PMID: 37315833 DOI: 10.1016/j.exer.2023.109539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
Alkali burn-induced corneal injury often causes inflammation and neovascularization and leads to compromised vision. We previously reported that rapamycin ameliorated corneal injury after alkali burns by methylation modification. In this study, we aimed to investigate the rapamycin-medicated mechanism against corneal inflammation and neovascularization. Our data showed that alkali burn could induce a range of different inflammatory response, including a stark upregulation of pro-inflammatory factor expression and an increase in the infiltration of myeloperoxidase- and F4/80-positive cells from the corneal limbus to the central stroma. Rapamycin effectively downregulated the mRNA expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), toll-like receptor 4 (TLR4), nucleotide binding oligomerization domain-like receptors (NLR) family pyrin domain-containing 3 (NLRP3), and Caspase-1, and suppressed the infiltration of neutrophils and macrophages. Inflammation-related angiogenesis mediated by matrix metalloproteinase-2 (MMP-2) and rapamycin restrained this process by inhibiting the TNF-α upregulation in burned corneas of mice. Rapamycin also restrained corneal alkali burn-induced inflammation by regulating HIF-1α/VEGF-mediated angiogenesis and the serum cytokines TNF-α, IL-6, Interferon-gamma (IFN-γ) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The findings of this study indicated rapamycin may reduce inflammation-associated infiltration of inflammatory cells, shape the expression of cytokines, and balance the regulation of MMP-2 and HIF-1α-mediated inflammation and angiogenesis by suppressing mTOR activation in corneal wound healing induced by an alkali injury. It offered novel insights relevant for a potent drug for treating corneal alkali burn.
Collapse
Affiliation(s)
- Jiande Li
- College of Life Sciences, Northwest Normal University, Lanzhou, 730070, Gansu, China.
| | - Jiangyuan Han
- College of Life Sciences and Technology, Gansu Agricultural University, Lanzhou, 730070, Gansu, China.
| | - Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Minrui Liu
- College of Life Sciences and Technology, Gansu Agricultural University, Lanzhou, 730070, Gansu, China.
| |
Collapse
|
9
|
Shi G, Chiramel AI, Li T, Lai KK, Kenney AD, Zani A, Eddy AC, Majdoul S, Zhang L, Dempsey T, Beare PA, Kar S, Yewdell JW, Best SM, Yount JS, Compton AA. Rapalogs downmodulate intrinsic immunity and promote cell entry of SARS-CoV-2. J Clin Invest 2022; 132:e160766. [PMID: 36264642 PMCID: PMC9753997 DOI: 10.1172/jci160766] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/18/2022] [Indexed: 12/24/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in immunocompromised individuals is associated with prolonged virus shedding and evolution of viral variants. Rapamycin and its analogs (rapalogs, including everolimus, temsirolimus, and ridaforolimus) are FDA approved as mTOR inhibitors for the treatment of human diseases, including cancer and autoimmunity. Rapalog use is commonly associated with an increased susceptibility to infection, which has been traditionally explained by impaired adaptive immunity. Here, we show that exposure to rapalogs increased susceptibility to SARS-CoV-2 infection in tissue culture and in immunologically naive rodents by antagonizing the cell-intrinsic immune response. We identified 1 rapalog (ridaforolimus) that was less potent in this regard and demonstrated that rapalogs promote spike-mediated entry into cells, by triggering the degradation of the antiviral proteins IFITM2 and IFITM3 via an endolysosomal remodeling program called microautophagy. Rapalogs that increased virus entry inhibited mTOR-mediated phosphorylation of the transcription factor TFEB, which facilitated its nuclear translocation and triggered microautophagy. In rodent models of infection, injection of rapamycin prior to and after virus exposure resulted in elevated SARS-CoV-2 replication and exacerbated viral disease, while ridaforolimus had milder effects. Overall, our findings indicate that preexisting use of certain rapalogs may elevate host susceptibility to SARS-CoV-2 infection and disease by activating lysosome-mediated suppression of intrinsic immunity.
Collapse
Affiliation(s)
- Guoli Shi
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Abhilash I. Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Hamilton, Montana, USA
| | - Tiansheng Li
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Kin Kui Lai
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Ashley Zani
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Adrian C. Eddy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Saliha Majdoul
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Lizhi Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Tirhas Dempsey
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| | - Paul A. Beare
- Laboratory of Bacteriology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | | | | | - Sonja M. Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Hamilton, Montana, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Alex A. Compton
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Frederick, Maryland, USA
| |
Collapse
|
10
|
Vellasamy DM, Lee SJ, Goh KW, Goh BH, Tang YQ, Ming LC, Yap WH. Targeting Immune Senescence in Atherosclerosis. Int J Mol Sci 2022; 23:13059. [PMID: 36361845 PMCID: PMC9658319 DOI: 10.3390/ijms232113059] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 10/29/2023] Open
Abstract
Atherosclerosis is one of the main underlying causes of cardiovascular diseases (CVD). It is associated with chronic inflammation and intimal thickening as well as the involvement of multiple cell types including immune cells. The engagement of innate or adaptive immune response has either athero-protective or atherogenic properties in exacerbating or alleviating atherosclerosis. In atherosclerosis, the mechanism of action of immune cells, particularly monocytes, macrophages, dendritic cells, and B- and T-lymphocytes have been discussed. Immuno-senescence is associated with aging, viral infections, genetic predispositions, and hyperlipidemia, which contribute to atherosclerosis. Immune senescent cells secrete SASP that delays or accelerates atherosclerosis plaque growth and associated pathologies such as aneurysms and coronary artery disease. Senescent cells undergo cell cycle arrest, morphological changes, and phenotypic changes in terms of their abundances and secretome profile including cytokines, chemokines, matrix metalloproteases (MMPs) and Toll-like receptors (TLRs) expressions. The senescence markers are used in therapeutics and currently, senolytics represent one of the emerging treatments where specific targets and clearance of senescent cells are being considered as therapy targets for the prevention or treatment of atherosclerosis.
Collapse
Affiliation(s)
- Danusha Michelle Vellasamy
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Sin-Jye Lee
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yin-Quan Tang
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| |
Collapse
|
11
|
Richman S, Lyman C, Nesterova A, Yuryev A, Morris M, Cao H, Cheadle C, Skuse G, Broderick G. Old drugs, new tricks: leveraging known compounds to disrupt coronavirus-induced cytokine storm. NPJ Syst Biol Appl 2022; 8:38. [PMID: 36216820 PMCID: PMC9549818 DOI: 10.1038/s41540-022-00250-9] [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: 02/04/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022] Open
Abstract
A major complication in COVID-19 infection consists in the onset of acute respiratory distress fueled by a dysregulation of the host immune network that leads to a run-away cytokine storm. Here, we present an in silico approach that captures the host immune system's complex regulatory dynamics, allowing us to identify and rank candidate drugs and drug pairs that engage with minimal subsets of immune mediators such that their downstream interactions effectively disrupt the signaling cascades driving cytokine storm. Drug-target regulatory interactions are extracted from peer-reviewed literature using automated text-mining for over 5000 compounds associated with COVID-induced cytokine storm and elements of the underlying biology. The targets and mode of action of each compound, as well as combinations of compounds, were scored against their functional alignment with sets of competing model-predicted optimal intervention strategies, as well as the availability of like-acting compounds and known off-target effects. Top-ranking individual compounds identified included a number of known immune suppressors such as calcineurin and mTOR inhibitors as well as compounds less frequently associated for their immune-modulatory effects, including antimicrobials, statins, and cholinergic agonists. Pairwise combinations of drugs targeting distinct biological pathways tended to perform significantly better than single drugs with dexamethasone emerging as a frequent high-ranking companion. While these predicted drug combinations aim to disrupt COVID-induced acute respiratory distress syndrome, the approach itself can be applied more broadly to other diseases and may provide a standard tool for drug discovery initiatives in evaluating alternative targets and repurposing approved drugs.
Collapse
Affiliation(s)
- Spencer Richman
- Rochester General Hospital, Center for Clinical Systems Biology, Rochester, NY, USA
| | - Cole Lyman
- Rochester General Hospital, Center for Clinical Systems Biology, Rochester, NY, USA
| | | | - Anton Yuryev
- Elsevier BV, Biology Solutions, Amsterdam, the Netherlands
| | - Matthew Morris
- Rochester General Hospital, Center for Clinical Systems Biology, Rochester, NY, USA
| | - Hongbao Cao
- Elsevier BV, Biology Solutions, Amsterdam, the Netherlands
| | - Chris Cheadle
- Elsevier BV, Biology Solutions, Amsterdam, the Netherlands
| | - Gary Skuse
- Rochester Institute of Technology, Gosnell School of Life Sciences, Rochester, NY, USA
| | - Gordon Broderick
- Rochester General Hospital, Center for Clinical Systems Biology, Rochester, NY, USA.
| |
Collapse
|
12
|
Marrella V, Facoetti A, Cassani B. Cellular Senescence in Immunity against Infections. Int J Mol Sci 2022; 23:11845. [PMID: 36233146 PMCID: PMC9570409 DOI: 10.3390/ijms231911845] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is characterized by irreversible cell cycle arrest in response to different triggers and an inflammatory secretome. Although originally described in fibroblasts and cell types of solid organs, cellular senescence affects most tissues with advancing age, including the lymphoid tissue, causing chronic inflammation and dysregulation of both innate and adaptive immune functions. Besides its normal occurrence, persistent microbial challenge or pathogenic microorganisms might also accelerate the activation of cellular aging, inducing the premature senescence of immune cells. Therapeutic strategies counteracting the detrimental effects of cellular senescence are being developed. Their application to target immune cells might have the potential to improve immune dysfunctions during aging and reduce the age-dependent susceptibility to infections. In this review, we discuss how immune senescence influences the host's ability to resolve more common infections in the elderly and detail the different markers proposed to identify such senescent cells; the mechanisms by which infectious agents increase the extent of immune senescence are also reviewed. Finally, available senescence therapeutics are discussed in the context of their effects on immunity and against infections.
Collapse
Affiliation(s)
- Veronica Marrella
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, 20138 Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Amanda Facoetti
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Barbara Cassani
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, Università Degli Studi di Milano, 20089 Milan, Italy
| |
Collapse
|
13
|
The Mycophenolate-based Immunosuppressive Regimen Is Associated With Increased Mortality in Kidney Transplant Patients With COVID-19. Transplantation 2022; 106:e441-e451. [PMID: 35765133 PMCID: PMC9521389 DOI: 10.1097/tp.0000000000004251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The chronic use of immunosuppressive drugs is a key risk factor of death because of coronavirus disease 2019 (COVID-19) in kidney transplant recipients (KTRs), although no evident association between the class of immunosuppressive and outcomes has been observed. Thus, we aimed to compare COVID-19-associated outcomes among KTRs receiving 3 different immunosuppressive maintenance regimes. METHODS This study included data from 1833 KTRs with COVID-19 diagnosed between March 20 and April 21 extracted from the national registry before immunization. All patients were taking calcineurin inhibitor associated with mycophenolate acid (MPA, n = 1258), azathioprine (AZA, n = 389), or mammalian targets of rapamycin inhibitors (mTORi, n = 186). Outcomes within 30 and 90 d were assessed. RESULTS Compared with patients receiving MPA, the 30-d (79.9% versus 87.9% versus 89.2%; P < 0.0001) and 90-d (75% versus 83.5% versus 88.2%; P < 0.0001) unadjusted patient survivals were higher in those receiving AZA or mTORi, respectively. Using adjusted multivariable Cox regression, compared with patients receiving AZA, the use of MPA was associated with a higher risk of death within 30 d (adjusted hazard ratio [aHR], 1.70; 95% confidence interval [CI], 1.21-2.40; P = 0.003), which was not observed in patients using mTORi (aHR, 0.78; 95% CI, 0.45-1.35; P = 0.365). At 90 d, although higher risk of death was confirmed in patients receiving MPA (aHR, 1.46; 95% CI, 1.09-1.98; P = 0.013), a reduced risk was observed in patients receiving mTORi (aHR, 0.59; 95% CI, 0.35-0.97; P = 0.04) compared with AZA. CONCLUSIONS This national cohort data suggest that, in KTRs receiving calcineurin inhibitor and diagnosed with COVID-19, the use of MPA was associated with higher risk of death, whereas mTORi use was associated with lower risk of death.
Collapse
|
14
|
Jiang Y, Zhao T, Zhou X, Xiang Y, Gutierrez‐Castrellon P, Ma X. Inflammatory pathways in COVID-19: Mechanism and therapeutic interventions. MedComm (Beijing) 2022; 3:e154. [PMID: 35923762 PMCID: PMC9340488 DOI: 10.1002/mco2.154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
The 2019 coronavirus disease (COVID-19) pandemic has become a global crisis. In the immunopathogenesis of COVID-19, SARS-CoV-2 infection induces an excessive inflammatory response in patients, causing an inflammatory cytokine storm in severe cases. Cytokine storm leads to acute respiratory distress syndrome, pulmonary and other multiorgan failure, which is an important cause of COVID-19 progression and even death. Among them, activation of inflammatory pathways is a major factor in generating cytokine storms and causing dysregulated immune responses, which is closely related to the severity of viral infection. Therefore, elucidation of the inflammatory signaling pathway of SARS-CoV-2 is important in providing otential therapeutic targets and treatment strategies against COVID-19. Here, we discuss the major inflammatory pathways in the pathogenesis of COVID-19, including induction, function, and downstream signaling, as well as existing and potential interventions targeting these cytokines or related signaling pathways. We believe that a comprehensive understanding of the regulatory pathways of COVID-19 immune dysregulation and inflammation will help develop better clinical therapy strategies to effectively control inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Yujie Jiang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Xueyan Zhou
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Yu Xiang
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| | - Pedro Gutierrez‐Castrellon
- Center for Translational Research on Health Science Hospital General Dr. Manuel Gea GonzalezMinistry of HealthMexico CityMexico
| | - Xuelei Ma
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| |
Collapse
|
15
|
Shi G, Chiramel AI, Li T, Lai KK, Kenney AD, Zani A, Eddy A, Majdoul S, Zhang L, Dempsey T, Beare PA, Kar S, Yewdell JW, Best SM, Yount JS, Compton AA. Rapalogs downmodulate intrinsic immunity and promote cell entry of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.04.15.440067. [PMID: 33880473 PMCID: PMC8057238 DOI: 10.1101/2021.04.15.440067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SARS-CoV-2 infection in immunocompromised individuals is associated with prolonged virus shedding and evolution of viral variants. Rapamycin and its analogs (rapalogs, including everolimus, temsirolimus, and ridaforolimus) are FDA-approved as mTOR inhibitors for the treatment of human diseases, including cancer and autoimmunity. Rapalog use is commonly associated with increased susceptibility to infection, which has been traditionally explained by impaired adaptive immunity. Here, we show that exposure to rapalogs increases susceptibility to SARS-CoV-2 infection in tissue culture and in immunologically naive rodents by antagonizing the cell-intrinsic immune response. By identifying one rapalog (ridaforolimus) that is less potent in this regard, we demonstrate that rapalogs promote Spike-mediated entry into cells by triggering the degradation of antiviral proteins IFITM2 and IFITM3 via an endolysosomal remodeling program called microautophagy. Rapalogs that increase virus entry inhibit the mTOR-mediated phosphorylation of the transcription factor TFEB, which facilitates its nuclear translocation and triggers microautophagy. In rodent models of infection, injection of rapamycin prior to and after virus exposure resulted in elevated SARS-CoV-2 replication and exacerbated viral disease, while ridaforolimus had milder effects. Overall, our findings indicate that preexisting use of certain rapalogs may elevate host susceptibility to SARS-CoV-2 infection and disease by activating lysosome-mediated suppression of intrinsic immunity.
Collapse
Affiliation(s)
- Guoli Shi
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Abhilash I. Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Tiansheng Li
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Kin Kui Lai
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Ashley Zani
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Adrian Eddy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Saliha Majdoul
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Lizhi Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Tirhas Dempsey
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Paul A. Beare
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | | | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Sonja M. Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Alex A. Compton
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
16
|
Targeting autophagy regulation in NLRP3 inflammasome-mediated lung inflammation in COVID-19. Clin Immunol 2022; 244:109093. [PMID: 35944881 PMCID: PMC9356669 DOI: 10.1016/j.clim.2022.109093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Emerging evidence indicates that the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome is activated, which results in a cytokine storm at the late stage of COVID-19. Autophagy regulation is involved in the infection and replication of SARS-CoV-2 at the early stage and the inhibition of NLRP3 inflammasome-mediated lung inflammation at the late stage of COVID-19. Here, we discuss the autophagy regulation at different stages of COVID-19. Specifically, we highlight the therapeutic potential of autophagy activators in COVID-19 by inhibiting the NLRP3 inflammasome, thereby avoiding the cytokine storm. We hope this review provides enlightenment for the use of autophagy activators targeting the inhibition of the NLRP3 inflammasome, specifically the combinational therapy of autophagy modulators with the inhibitors of the NLRP3 inflammasome, antiviral drugs, or anti-inflammatory drugs in the fight against COVID-19.
Collapse
|
17
|
Attia MH. A cautionary note on altered pace of aging in the COVID-19 era. Forensic Sci Int Genet 2022; 59:102724. [PMID: 35598567 PMCID: PMC9112667 DOI: 10.1016/j.fsigen.2022.102724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/18/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is highly age-dependent due to hi-jacking the molecular control of the immune cells by the severe acute respiratory syndrome-corona virus 2 (SARS-CoV-2) leading to aberrant DNA methylation (DNAm) pattern of blood in comparison to normal individuals. These epigenetic modifications have been linked to perturbations to the epigenetic clock, development of long COVID-19 syndrome, and all-cause mortality risk. I reviewed the effects of COVID-19 on different molecular age markers such as the DNAm, telomere length (TL), and signal joint T-cell receptor excision circle (sjTREC). Integrating the accumulated clinical research data, COVID-19 and novel medical management may alter the pace of aging in adult individuals (<60 years). As such, COVID-19 might be a confounder in epigenetic age estimation similar to life style diversities, pathogens and pathologies which may influence the interpretation of DNAm data. Similarly, the SARS-CoV-2 affects T-lymphocyte function with possible influence on sjTREC levels. In contrast, TL measurements performed years before the SARS-CoV-2 pandemic proved that short TL predisposes to severe COVID- 19 independently from chronological age. However, the persistence of COVID-19 epigenetic scars and the durability of the immune response after vaccination and their effect on the ongoing pace of aging are still unknown. In the light of these data, the heterogeneous nature of the samples in these studies mandates a systematic evaluation of the currrent methods. SARS-CoV-2 may modify the reliability of the age estimation models in real casework because blood is the most common biological sample encountered in forensic contexts.
Collapse
|
18
|
Zalpoor H, Bakhtiyari M, Liaghat M, Nabi‐Afjadi M, Ganjalikhani‐Hakemi M. Quercetin potential effects against SARS-CoV-2 infection and COVID-19-associated cancer progression by inhibiting mTOR and hypoxia-inducible factor-1α (HIF-1α). Phytother Res 2022; 36:2679-2682. [PMID: 35307904 PMCID: PMC9111038 DOI: 10.1002/ptr.7440] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023]
Affiliation(s)
- Hamidreza Zalpoor
- American Association of Kidney PatientsTampaFloridaUSA
- Shiraz Neuroscience Research CenterShiraz University of Medical SciencesShirazIran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN)TehranIran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Medical Laboratory Sciences, Faculty of Allied MedicineQazvin University of Medical SciencesQazvinIran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Medical Laboratory sciences, Faculty of Medical Sciences, Kazerun BranchIslamic Azad UniversityKazerunIran
| | - Mohsen Nabi‐Afjadi
- Department of Biochemistry, Faculty of Biological ScienceTarbiat Modares UniversityTehranIran
| | - Mazdak Ganjalikhani‐Hakemi
- Department of Immunology, Faculty of MedicineIsfahan University of Medical SciencesIsfahanIran
- Acquired Immunodeficiency Research CenterIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
19
|
Mohamed MA, Elkhateeb WA, Daba GM. Rapamycin golden jubilee and still the miraculous drug: a potent immunosuppressant, antitumor, rejuvenative agent, and potential contributor in COVID-19 treatment. BIORESOUR BIOPROCESS 2022; 9:65. [PMID: 35730039 PMCID: PMC9188914 DOI: 10.1186/s40643-022-00554-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
Although celebrating its golden jubilee, rapamycin’s importance keeps increasing by the day. Starting as a promising antifungal agent, then as a potent immunosuppressant, strong anticancer drug, and now rapamycin is attracting serious attention as a rejuvenative agent and a possible contributor in treating this era pandemic, COVID-19. Due to its diverse biological activities and promising medical applications, we aimed in this review to put rapamycin under the spot and highlight its discovery, famous microbial producers, reported biological activities, chemical structure, famous analogues, and biosynthesis. Moreover, discuss some rapamycin production approaches including solid-state fermentation, and stressing out producing strain. On the other hand, describe its action mechanism and trials to use it in treatment of COVID-19. Additionally, we highlighted some of the side effects accompanying its use, and describe some approaches reported to minimize these undesired effects. Finally, we report the current status of rapamycin and its analogues in global market, and discuss future prospects of this potent drug.
Collapse
Affiliation(s)
- Mohamed A Mohamed
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| | - Waill A Elkhateeb
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| | - Ghoson M Daba
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| |
Collapse
|
20
|
Pinchera B, Spirito L, Buonomo AR, Foggia M, Carrano R, Salemi F, Schettino E, Papa F, La Rocca R, Crocetto F, Napolitano L, Villari R, Gentile I. mTOR Inhibitor Use Is Associated With a Favorable Outcome of COVID-19 in Patients of Kidney Transplant: Results of a Retrospective Study. Front Med (Lausanne) 2022; 9:852973. [PMID: 35801204 PMCID: PMC9254357 DOI: 10.3389/fmed.2022.852973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/29/2022] [Indexed: 11/22/2022] Open
Abstract
Introduction In solid organ transplant recipients, COVID-19 is associated with a poor prognosis because of immunosuppression. Some studies suggest a potential therapeutic role of mammalian Target of Rapamycin (mTOR) inhibitors in SARS-CoV-2 infection. This study aimed to assess the impact of mTOR employment on the evolution and outcome of SARS-CoV-2 infection in solid organ transplant recipients. Methods We enrolled kidney transplant patients attending the Azienda Ospedaliera Universitaria Federico II in Naples and followed up on these patients from March 2020 to June 2021. We evaluated the risk of acquiring the SARS-CoV-2 infection, the clinical presentation of the disease, and its outcome together with the type of immunosuppressive therapy. Finally, we assessed the impact of mTOR inhibitors on relevant clinical metrics of SARS-CoV-2 infection. Results We enrolled 371 patients, of whom 56 (15.1%) contracted SARS-CoV-2 infection during the period of the study. There were no differences observed among the different immunosuppressive therapies concerning the risk of acquiring SARS-CoV-2 infection. In contrast, the type of immunosuppressive therapy had a significant impact on the outcome of the disease. In detail, patients who received mTOR inhibitors, as part of their immunosuppressive therapy, compared to other regimens had a lower chance of developing a moderate or severe form of the disease (OR = 0.8, 95, CI: (0.21-0.92), P = 0.041). Conclusion In kidney transplant patients, the use of mTOR inhibitors as part of an immunosuppressive regimen is associated with a better prognosis in the case of COVID-19.
Collapse
Affiliation(s)
- Biagio Pinchera
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lorenzo Spirito
- Section of Urology, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Riccardo Buonomo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Foggia
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rosa Carrano
- Section of Nephrology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabrizio Salemi
- Section of Nephrology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Elisa Schettino
- Section of Nephrology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fortuna Papa
- Section of Nephrology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Roberto La Rocca
- Section of Urology, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Felice Crocetto
- Section of Urology, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Luigi Napolitano
- Section of Urology, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Riccardo Villari
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ivan Gentile
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
21
|
Mortezaee K, Majidpoor J. CD8 + T Cells in SARS-CoV-2 Induced Disease and Cancer-Clinical Perspectives. Front Immunol 2022; 13:864298. [PMID: 35432340 PMCID: PMC9010719 DOI: 10.3389/fimmu.2022.864298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Dysregulated innate and adaptive immunity is a sign of SARS-CoV-2-induced disease and cancer. CD8+ T cells are important cells of the immune system. The cells belong to the adaptive immunity and take a front-line defense against viral infections and cancer. Extreme CD8+ T-cell activities in the lung of patients with a SARS-CoV-2-induced disease and within the tumor microenvironment (TME) will change their functionality into exhausted state and undergo apoptosis. Such diminished immunity will put cancer cases at a high-risk group for SARS-CoV-2-induced disease, rendering viral sepsis and a more severe condition which will finally cause a higher rate of mortality. Recovering responses from CD8+ T cells is a purpose of vaccination against SARS-CoV-2. The aim of this review is to discuss the CD8+ T cellular state in SARS-CoV-2-induced disease and in cancer and to present some strategies for recovering the functionality of these critical cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
22
|
Kumari M, Kumar A. Environmental and human health risk assessment of mixture of Covid-19 treating pharmaceutical drugs in environmental waters. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 812:152485. [PMID: 34942257 PMCID: PMC8686450 DOI: 10.1016/j.scitotenv.2021.152485] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 04/14/2023]
Abstract
This study identified ecological and human health risks exposure of COVID-19 pharmaceuticals and their metabolites in environmental waters. Environmental concentrations in aquatic species were predicted using surface water concentrations of pharmaceutical compounds. Predicted No-Effect Concentrations (PNEC) in aquatic organisms (green algae, daphnia, and fish) was estimated using EC50/LC50 values of pharmaceutical compounds taken from USEPA ECOSAR database. PNEC for human health risks was calculated using the acceptable daily intake values of drugs. Ecological PNEC revealed comparatively high values in algae (Chronic toxicity PNEC values, high to low: ribavirin (2.65 × 105 μg/L) to ritonavir (2.3 × 10-1 μg/L)) than daphnia and fish. Risk quotient (RQ) analysis revealed that algae (Avg. = 2.81 × 104) appeared to be the most sensitive species to pharmaceutical drugs followed by daphnia (Avg.: 1.28 × 104) and fish (Avg.: 1.028 × 103). Amongst the COVID-19 metabolites, lopinavir metabolites posed major risk to aquatic species. Ritonavir (RQ = 6.55) is the major drug responsible for human health risk through consumption of food (in the form fish) grown in pharmaceutically contaminated waters. Mixture toxicity analysis of drugs revealed that algae are the most vulnerable species amongst the three trophic levels. Maximum allowable concentration level for mixture of pharmaceuticals was found to be 0.53 mg/L.
Collapse
Affiliation(s)
- Minashree Kumari
- Environment Engineering Section, Department of Civil Engineering, Indian Institute of Technology Delhi, Hauz Khas, Delhi, 110017, India.
| | - Arun Kumar
- Environment Engineering Section, Department of Civil Engineering, Indian Institute of Technology Delhi, Hauz Khas, Delhi, 110017, India.
| |
Collapse
|
23
|
Gauthier T, Chen W. Modulation of Macrophage Immunometabolism: A New Approach to Fight Infections. Front Immunol 2022; 13:780839. [PMID: 35154105 PMCID: PMC8825490 DOI: 10.3389/fimmu.2022.780839] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Macrophages are essential innate immune cells that contribute to host defense during infection. An important feature of macrophages is their ability to respond to extracellular cues and to adopt different phenotypes and functions in response to these stimuli. The evidence accumulated in the last decade has highlighted the crucial role of metabolic reprogramming during macrophage activation in infectious context. Thus, understanding and manipulation of macrophage immunometabolism during infection could be of interest to develop therapeutic strategies. In this review, we focus on 5 major metabolic pathways including glycolysis, pentose phosphate pathway, fatty acid oxidation and synthesis, tricarboxylic acid cycle and amino acid metabolism and discuss how they sustain and regulate macrophage immune function in response to parasitic, bacterial and viral infections as well as trained immunity. At the end, we assess whether some drugs including those used in clinic and in development can target macrophage immunometabolism for potential therapy during infection with an emphasis on SARS-CoV2 infection.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wanjun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
24
|
Morgulchik N, Athanasopoulou F, Chu E, Lam Y, Kamaly N. Potential therapeutic approaches for targeted inhibition of inflammatory cytokines following COVID-19 infection-induced cytokine storm. Interface Focus 2022; 12:20210006. [PMID: 34956607 PMCID: PMC8662389 DOI: 10.1098/rsfs.2021.0006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a deadly respiratory disease caused by severe acute respiratory syndrome coronavirus 2, which has caused a global pandemic since early 2020 and severely threatened people's livelihoods and health. Patients with pre-diagnosed conditions admitted to hospital often develop complications leading to mortality due to acute respiratory distress syndrome (ARDS) and associated multiorgan failure and blood clots. ARDS is associated with a cytokine storm. Cytokine storms arise due to elevated levels of circulating cytokines and are associated with infections. Targeting various pro-inflammatory cytokines in a specific manner can result in a potent therapeutic approach with minimal host collateral damage. Immunoregulatory therapies are now of interest in order to regulate the cytokine storm, and this review will summarize and discuss advances in targeted therapies against cytokine storms induced by COVID-19.
Collapse
Affiliation(s)
- Nelli Morgulchik
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Foteini Athanasopoulou
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Edmund Chu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Yoriko Lam
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| |
Collapse
|
25
|
Fattahi S, Khalifehzadeh-Esfahani Z, Mohammad-Rezaei M, Mafi S, Jafarinia M. PI3K/Akt/mTOR pathway: a potential target for anti-SARS-CoV-2 therapy. Immunol Res 2022; 70:269-275. [PMID: 35107743 PMCID: PMC8808470 DOI: 10.1007/s12026-022-09268-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/30/2022] [Indexed: 02/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A single-stranded RNA virus from a β-Coronaviridae family causes acute clinical manifestations. Its high death rate and severe clinical symptoms have turned it into the most significant challenge worldwide. Up until now, several effective COVID-19 vaccines have been designed and marketed, but our data on specialized therapeutic drugs for the treatment of COVID-19 is still limited. In order to synthesis virus particles, SARS-CoV-2 uses host metabolic pathways such as phosphoinositide3-kinase (PI3K)/protein kinase B (PKB, also known as AKT)/mammalian target of rapamycin (mTOR). mTOR is involved in multiple biological processes. Over-activation of the mTOR pathway improves viral replication, which makes it a possible target in COVID-19 therapy. Clinical data shows the hyperactivation of the mTOR pathway in lung tissues during respiratory viral infections. However, the exact impact of mTOR pathway inhibitors on the COVID-19 severity and death rate is yet to be thoroughly investigated. There are several mTOR pathway inhibitors. Rapamycin is the most famous inhibitor of mTORC1 among all. Studies on other respiratory viruses suggest that the therapeutic inhibitors of the mTOR pathway, especially rapamycin, can be a potential approach to anti-SARS-CoV-2 therapy. Using therapeutic methods that inhibit harmful immune responses can open a new chapter in treating severe COVID-19 disease. We highlighted the potential contribution of PI3K/Akt/mTOR inhibitors in the treatment of COVID-19.
Collapse
Affiliation(s)
- Soheila Fattahi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Mina Mohammad-Rezaei
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sahar Mafi
- Medical Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
26
|
Blocking TNF signaling may save lives in COVID-19 infection. Mol Biol Rep 2022; 49:2303-2309. [PMID: 35076845 PMCID: PMC8787182 DOI: 10.1007/s11033-022-07166-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 12/30/2022]
Abstract
Global vaccination effort and better understanding of treatment strategies provided a ray of hope for improvement in COVID-19 pandemic, however, in many countries, the disease continues to collect its death toll. The major pathogenic mechanism behind severe cases associated with high mortality is the burst of pro-inflammatory cytokines TNF, IL-6, IFNγ and others, resulting in multiple organ failure. Although the exact contribution of each cytokine is not clear, we provide an evidence that the central mediator of cytokine storm and its devastating consequences may be TNF. This cytokine is known to be involved in activated blood clotting, lung damage, insulin resistance, heart failure, and other conditions. A number of currently available pharmaceutical agents such as monoclonal antibodies and soluble TNF receptors can effectively prevent TNF from binding to its receptor(s). Other drugs are known to block NFkB, the major signal transducer molecule used in TNF signaling, or to block kinases involved in downstream activation cascades. Some of these medicines have already been selected for clinical trials, but more work is needed. A simple, rapid, and inexpensive method of directly monitoring TNF levels may be a valuable tool for a timely selection of COVID-19 patients for anti-TNF therapy.
Collapse
|
27
|
Karastaneva A, Gasparella P, Tschauner S, Crazzolara R, Kropshofer G, Modl M, Pfleger A, Burmas A, Pocivalnik M, Ulreich R, Zenz W, Schwinger W, Beqo BP, Urban C, Haxhija EQ, Lackner H, Benesch M. Indications and Limitations of Sirolimus in the Treatment of Vascular Anomalies-Insights From a Retrospective Case Series. Front Pediatr 2022; 10:857436. [PMID: 35676905 PMCID: PMC9168223 DOI: 10.3389/fped.2022.857436] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Despite recent developments, the role of sirolimus in the heterogeneous spectrum of vascular anomalies is yet to be defined, in terms of indication, dosage, and therapy duration, recognizing both its potential and limitations. METHODS We retrospectively analyzed 16 children with vascular anomalies treated with sirolimus in two pediatric centers between 2014 and 2020 [male: n = 7, the median age at diagnosis: 4.6 months (range, 0-281.4)]. In addition, repetitive volumetric analyses of the vascular anomalies were performed when possible (11 cases). RESULTS Ten patients were diagnosed with vascular malformations and 6 with vascular tumors. The mean therapy duration was 27.2 months (range, 3.5-65). The mean sirolimus level was 8.52 ng/ml (range, 5.38-12.88). All patients except one with central conducting lymphatic anomaly responded to sirolimus, with the most noticeable volume reduction in the first 4-6 months. Additional administration of vincristine was needed in five patients with kaposiform hemangioendothelioma and yielded a response, even in cases, refractory to sirolimus monotherapy. As a single agent, sirolimus led to impressive improvement in a patient with another vascular tumor-advanced epithelioid hemangioendothelioma. Complicated vascular malformations required long-term sirolimus therapy. Side effects of sirolimus included mucositis and laboratory abnormalities. No major infectious episodes were recorded. An infant with COVID-19, diagnosed while on sirolimus therapy, presented with a mild course. CONCLUSION In the current series, we reported limitations of sirolimus as monotherapy, addressing the need to redefine its indications, and explore combination regimens and multimodal treatment strategies. Tools for objective evaluation of response trends over time could serve as a basis for the establishment of future therapeutic algorithms.
Collapse
Affiliation(s)
- Anna Karastaneva
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Paolo Gasparella
- Department of Pediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| | | | - Roman Crazzolara
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Kropshofer
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Modl
- Division of Pediatric Pulmonology and Allergology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Pfleger
- Division of Pediatric Pulmonology and Allergology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Ante Burmas
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Mirjam Pocivalnik
- Pediatric Intensive Care Unit, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Raphael Ulreich
- Pediatric Intensive Care Unit, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Werner Zenz
- Division of General Pediatrics, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Wolfgang Schwinger
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Besiana P Beqo
- Department of Pediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria.,Global Clinical Scholars Research Training, Department of Postgraduate Medical Education, Harvard Medical School, Boston, MA, United States
| | - Christian Urban
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Emir Q Haxhija
- Department of Pediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| | - Herwig Lackner
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Martin Benesch
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
28
|
Farheen S, Agrawal S, Zubair S, Agrawal A, Jamal F, Altaf I, Kashif Anwar A, Umair SM, Owais M. Patho-Physiology of Aging and Immune-Senescence: Possible Correlates With Comorbidity and Mortality in Middle-Aged and Old COVID-19 Patients. FRONTIERS IN AGING 2021; 2:748591. [PMID: 35822018 PMCID: PMC9261314 DOI: 10.3389/fragi.2021.748591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/30/2021] [Indexed: 01/08/2023]
Abstract
During the last 2 years, the entire world has been severely devastated by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic (COVID-19) as it resulted in several million deaths across the globe. While the virus infects people indiscriminately, the casualty risk is higher mainly in old, and middle-aged COVID-19 patients. The incidences of COVID-19 associated co-morbidity and mortality have a great deal of correlation with the weakened and malfunctioning immune systems of elderly people. Presumably, due to the physiological changes associated with aging and because of possible comorbidities such as diabetes, hypertension, obesity, cardiovascular, and lung diseases, which are more common in elderly people, may be considered as the reason making the elderly vulnerable to the infection on one hand, and COVID-19 associated complications on the other. The accretion of senescent immune cells not only contributes to the deterioration of host defense, but also results in elevated inflammatory phenotype persuaded immune dysfunction. In the present review, we envisage to correlate functioning of the immune defense of older COVID-19 patients with secondary/super infection, increased susceptibility or aggravation against already existing cancer, infectious, autoimmune, and other chronic inflammatory diseases. Moreover, we have discussed how age-linked modulations in the immune system affect therapeutic response against administered drugs as well as immunological response to various prophylactic measures including vaccination in the elderly host. The present review also provides an insight into the intricate pathophysiology of the aging and the overall immune response of the host to SARS-CoV-2 infection. A better understanding of age-related immune dysfunction is likely to help us in the development of targeted preemptive strategies for deadly COVID-19 in elderly patients.
Collapse
Affiliation(s)
- Saba Farheen
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Swaleha Zubair
- Department of Computer Science, Aligarh Muslim University, Aligarh, India
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Fauzia Jamal
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Ishrat Altaf
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Abu Kashif Anwar
- Department of Anatomy, HSZH Gov, Unani Medical College, Bhopal, India
| | | | - Mohammad Owais
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
- *Correspondence: Mohammad Owais,
| |
Collapse
|
29
|
Tuli HS, Sak K, Aggarwal P, Iqubal A, Upadhaya SK, Kaur J, Kaur G, Aggarwal D. Molecular Evolution of Severe Acute Respiratory Syndrome Coronavirus 2: Hazardous and More Hazardous Strains Behind the Coronavirus Disease 2019 Pandemic and Their Targeting by Drugs and Vaccines. Front Cell Infect Microbiol 2021; 11:763687. [PMID: 34970505 PMCID: PMC8712944 DOI: 10.3389/fcimb.2021.763687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Within almost the last 2 years, the world has been shaken by the coronavirus disease 2019 (COVID-19) pandemic, which has affected the lives of all people. With nearly 4.92 million deaths by October 19, 2021, and serious health damages in millions of people, COVID-19 has been the most serious global challenge after the Second World War. Besides lost lives and long-term health problems, devastating impact on economics, education, and culture will probably leave a lasting impression on the future. Therefore, the actual extent of losses will become obvious only after years. Moreover, despite the availability of different vaccines and vaccination programs, it is still impossible to forecast what the next steps of the virus are or how near we are to the end of the pandemic. In this article, the route of molecular evolution of the coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is thoroughly compiled, highlighting the changes that the virus has undergone during the last 2 years and discussing the approaches that the medical community has undertaken in the fight against virus-induced damages.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, India
| | - Katrin Sak
- Non-Governmental Organization (NGO) Praeventio, Tartu, Estonia
| | - Poonam Aggarwal
- The Basic Research Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, United States
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy), Jamia Hamdard (Deemed to be University), Delhi, India
| | - Sushil K. Upadhaya
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale BioPhotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Shri Vile Parle Kelavani Mandal, Narsee Monjee Institute of Management Studies (SVKM’S NMIMS), Mumbai, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, India
| |
Collapse
|
30
|
Wienecke LM, Cohen S, Bauersachs J, Mebazaa A, Chousterman BG. Immunity and inflammation: the neglected key players in congenital heart disease? Heart Fail Rev 2021; 27:1957-1971. [PMID: 34855062 PMCID: PMC8636791 DOI: 10.1007/s10741-021-10187-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/23/2022]
Abstract
Although more than 90% of children born with congenital heart disease (CHD) survive into adulthood, patients face significantly higher and premature morbidity and mortality. Heart failure as well as non-cardiac comorbidities represent a striking and life-limiting problem with need for new treatment options. Systemic chronic inflammation and immune activation have been identified as crucial drivers of disease causes and progression in various cardiovascular disorders and are promising therapeutic targets. Accumulating evidence indicates an inflammatory state and immune alterations in children and adults with CHD. In this review, we highlight the implications of chronic inflammation, immunity, and immune senescence in CHD. In this context, we summarize the impact of infant open-heart surgery with subsequent thymectomy on the immune system later in life and discuss the potential role of comorbidities and underlying genetic alterations. How an altered immunity and chronic inflammation in CHD influence patient outcomes facing SARS-CoV-2 infection is unclear, but requires special attention, as CHD could represent a population particularly at risk during the COVID-19 pandemic. Concluding remarks address possible clinical implications of immune changes in CHD and consider future immunomodulatory therapies.
Collapse
Affiliation(s)
- Laura M Wienecke
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30621, Hannover, Germany.
- Department of Anaesthesiology and Critical Care, Lariboisière University Hospital, DMU Parabol, AP-HP, Paris, France.
- Inserm U942 MASCOT, Université de Paris, Paris, France.
- Department of Cardiology, Angiology and Respiratory Medicine, Heidelberg University Hospital, Heidelberg, Germany.
| | - Sarah Cohen
- Congenital Heart Diseases Department, M3C Hospital Marie Lannelongue, Université Paris-Saclay, Plessis-Robinson, Paris, France
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30621, Hannover, Germany
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care, Lariboisière University Hospital, DMU Parabol, AP-HP, Paris, France
- Inserm U942 MASCOT, Université de Paris, Paris, France
| | - Benjamin G Chousterman
- Department of Anaesthesiology and Critical Care, Lariboisière University Hospital, DMU Parabol, AP-HP, Paris, France
- Inserm U942 MASCOT, Université de Paris, Paris, France
| |
Collapse
|
31
|
Erukainure OL, Atolani O, Muhammad A, Ravichandran R, Abarshi MM, Katsayal SB, Chukwuma CI, Preissner R, Banerjee P, Mesaik MA. Translational suppression of SARS-COV-2 ORF8 protein mRNA as a Viable therapeutic target against COVID-19: Computational studies on potential roles of isolated compounds from Clerodendrum volubile leaves. Comput Biol Med 2021; 139:104964. [PMID: 34688170 PMCID: PMC8524706 DOI: 10.1016/j.compbiomed.2021.104964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
The open reading frame 8 (ORF8) protein of SARS-CoV-2 has been implicated in the onset of cytokine storms, which are responsible for the pathophysiology of COVID-19 infection. The present study investigated the potential of isolated compounds from Clerodendrum volubile leaves to stall oxidative bursts in vitro and interact with ORF8 mRNA segments of the SARS-CoV-2 whole genome using computational tools. Five compounds, namely, harpagide, 1-(3-methyl-2-butenoxy)-4-(1-propenyl)benzene, ajugoside, iridoid glycoside and erucic acid, were isolated from C. volubile leaves, and their structures were elucidated using conventional spectroscopy tools. Iridoid glycoside is being reported for the first time and is thus regarded as a new compound. The ORF8 mRNA sequences of the translation initiation sites (TIS) and translation termination sites (TTSs) encoding ORF8 amino acids were retrieved from the full genome of SARS-CoV-2. Molecular docking studies revealed strong molecular interactions of the isolated compounds with the TIS and TTS of ORF8 mRNA. Harpagide showed the strongest binding affinity for TIS, while erucic acid was the strongest for TTS. The immunomodulatory potentials of the isolated compounds were investigated on neutrophil phagocytic respiratory bursts using luminol-amplified chemiluminescence technique. The compounds significantly inhibited oxidative burst, with 1-(3-methyl-2-butenoxy)-4-(1-propenyl)benzene having the best activity. Ajugoside and erucic acid showed significant inhibitory activity on T-cell proliferation. These results indicate the potential of C. volubile compounds as immunomodulators and can be utilized to curb cytokine storms implicated in COVID-19 infection. These potentials are further corroborated by the strong interactions of the compounds with the TIS and TTS of ORF8 mRNA from the SARS-CoV-2 whole genome.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa,Corresponding author
| | | | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Rahul Ravichandran
- DiSTABiF, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Musa M. Abarshi
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sanusi B. Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Chika I. Chukwuma
- Center for Quality of Health and Living, Faculty of Health Sciences, Central University of Technology, Bloemfontein 9301, South Africa
| | - Robert Preissner
- Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - Priyanka Banerjee
- Institute for Physiology, Charité – University Medicine Berlin, Berlin, Germany
| | - M. Ahmed Mesaik
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan,Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
32
|
Chiou WC, Hsu MS, Chen YT, Yang JM, Tsay YG, Huang HC, Huang C. Repurposing existing drugs: identification of SARS-CoV-2 3C-like protease inhibitors. J Enzyme Inhib Med Chem 2021; 36:147-153. [PMID: 33430659 PMCID: PMC7808739 DOI: 10.1080/14756366.2020.1850710] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/14/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for coronavirus disease 2019 (COVID-19). Since its emergence, the COVID-19 pandemic has not only distressed medical services but also caused economic upheavals, marking urgent the need for effective therapeutics. The experience of combating SARS-CoV and MERS-CoV has shown that inhibiting the 3-chymotrypsin-like protease (3CLpro) blocks the replication of the virus. Given the well-studied properties of FDA-approved drugs, identification of SARS-CoV-2 3CLpro inhibitors in an FDA-approved drug library would be of great therapeutic value. Here, we screened a library consisting of 774 FDA-approved drugs for potent SARS-CoV-2 3CLpro inhibitors, using an intramolecularly quenched fluorescence (IQF) peptide substrate. Ethacrynic acid, naproxen, allopurinol, butenafine hydrochloride, raloxifene hydrochloride, tranylcypromine hydrochloride, and saquinavir mesylate have been found to block the proteolytic activity of SARS-CoV-2 3CLpro. The inhibitory activity of these repurposing drugs against SARS-CoV-2 3CLpro highlights their therapeutic potential for treating COVID-19 and other Betacoronavirus infections.
Collapse
Affiliation(s)
- Wei-Chung Chiou
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Meng-Shiuan Hsu
- Departments of Infectious Disease, Far Eastern Memorial Hospital, Taipei, Taiwan
| | - Yun-Ti Chen
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu, Taiwan
- Faculty of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Yeou-Guang Tsay
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hsiu-Chen Huang
- Department of Applied Science, National Tsing Hua University South Campus, Hsinchu, Taiwan
| | - Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
33
|
Hu S, Jiang S, Qi X, Bai R, Ye XY, Xie T. Races of small molecule clinical trials for the treatment of COVID-19: An up-to-date comprehensive review. Drug Dev Res 2021; 83:16-54. [PMID: 34762760 PMCID: PMC8653368 DOI: 10.1002/ddr.21895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
The coronavirus disease‐19 (COVID‐19) pandemic has become a global threat since its first outbreak at the end of 2019. Several review articles have been published recently, focusing on the aspects of target biology, drug repurposing, and mechanisms of action (MOAs) for potential treatment. This review gathers all small molecules currently in active clinical trials, categorizes them into six sub‐classes, and summarizes their clinical progress. The aim is to provide the researchers from both pharmaceutical industries and academic institutes with the handful information and dataset to accelerate their research programs in searching effective small molecule therapy for treatment of COVID‐19.
Collapse
Affiliation(s)
- Suwen Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China.,Hangzhou Huadong Medicine Group, Pharmaceutical Research Institute Co. Ltd., Hangzhou, China.,Department of Chemistry and Biochemistry Los Angeles, University of California, Los Angeles, California, USA
| | - Songwei Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Xiang Qi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China.,Collaborative Innovation Center of Chinese Medicines from Zhejiang Province, Hangzhou Normal Umiversity, Hangzhou, China
| |
Collapse
|
34
|
Tomazou M, Bourdakou MM, Minadakis G, Zachariou M, Oulas A, Karatzas E, Loizidou EM, Kakouri AC, Christodoulou CC, Savva K, Zanti M, Onisiforou A, Afxenti S, Richter J, Christodoulou CG, Kyprianou T, Kolios G, Dietis N, Spyrou GM. Multi-omics data integration and network-based analysis drives a multiplex drug repurposing approach to a shortlist of candidate drugs against COVID-19. Brief Bioinform 2021; 22:bbab114. [PMID: 34009288 PMCID: PMC8135326 DOI: 10.1093/bib/bbab114] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/01/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is undeniably the most severe global health emergency since the 1918 Influenza outbreak. Depending on its evolutionary trajectory, the virus is expected to establish itself as an endemic infectious respiratory disease exhibiting seasonal flare-ups. Therefore, despite the unprecedented rally to reach a vaccine that can offer widespread immunization, it is equally important to reach effective prevention and treatment regimens for coronavirus disease 2019 (COVID-19). Contributing to this effort, we have curated and analyzed multi-source and multi-omics publicly available data from patients, cell lines and databases in order to fuel a multiplex computational drug repurposing approach. We devised a network-based integration of multi-omic data to prioritize the most important genes related to COVID-19 and subsequently re-rank the identified candidate drugs. Our approach resulted in a highly informed integrated drug shortlist by combining structural diversity filtering along with experts' curation and drug-target mapping on the depicted molecular pathways. In addition to the recently proposed drugs that are already generating promising results such as dexamethasone and remdesivir, our list includes inhibitors of Src tyrosine kinase (bosutinib, dasatinib, cytarabine and saracatinib), which appear to be involved in multiple COVID-19 pathophysiological mechanisms. In addition, we highlight specific immunomodulators and anti-inflammatory drugs like dactolisib and methotrexate and inhibitors of histone deacetylase like hydroquinone and vorinostat with potential beneficial effects in their mechanisms of action. Overall, this multiplex drug repurposing approach, developed and utilized herein specifically for SARS-CoV-2, can offer a rapid mapping and drug prioritization against any pathogen-related disease.
Collapse
Affiliation(s)
- Marios Tomazou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Marilena M Bourdakou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Greece
| | - George Minadakis
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Margarita Zachariou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Anastasis Oulas
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Evangelos Karatzas
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- Institute for Fundamental Biomedical Research, BSRC “Alexander Fleming”, Vari, Greece
| | - Eleni M Loizidou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- Institute for Fundamental Biomedical Research, BSRC “Alexander Fleming”, Vari, Greece
| | - Andrea C Kakouri
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Christiana C Christodoulou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
- Neuroepidemiology Department, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Kyriaki Savva
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Maria Zanti
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
- Cancer Genetics, Therapeutics … Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Anna Onisiforou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Sotiroula Afxenti
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
- Neuroimmunology Department, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - Jan Richter
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Christina G Christodoulou
- Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| | - Theodoros Kyprianou
- Medical School, University of Nicosia, Cyprus
- University Hospitals Bristol and Weston NHS Foundation Trust, United Kingdom
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Greece
| | - Nikolas Dietis
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, Cyprus
| | - George M Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Cyprus
- The Cyprus School of Molecular Medicine, Cyprus
| |
Collapse
|
35
|
Ying K, Zhai R, Pyrkov TV, Shindyapina AV, Mariotti M, Fedichev PO, Shen X, Gladyshev VN. Genetic and phenotypic analysis of the causal relationship between aging and COVID-19. COMMUNICATIONS MEDICINE 2021; 1:35. [PMID: 35602207 PMCID: PMC9053191 DOI: 10.1038/s43856-021-00033-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022] Open
Abstract
Background Epidemiological studies revealed that the elderly and those with comorbidities are most affected by COVID-19, but it is important to investigate shared genetic mechanisms between COVID-19 risk and aging. Methods We conducted a multi-instrument Mendelian Randomization analysis of multiple lifespan-related traits and COVID-19. Aging clock models were applied to the subjects with different COVID-19 conditions in the UK-Biobank cohort. We performed a bivariate genomic scan for age-related COVID-19 and Mendelian Randomization analysis of 389 immune cell traits to investigate their effect on lifespan and COVID-19 risk. Results We show that the genetic variation that supports longer life is significantly associated with the lower risk of COVID-19 infection and hospitalization. The odds ratio is 0.31 (P = 9.7 × 10-6) and 0.46 (P = 3.3 × 10-4), respectively, per additional 10 years of life. We detect an association between biological age acceleration and future incidence and severity of COVID-19 infection. Genetic profiling of age-related COVID-19 infection indicates key contributions of Notch signaling and immune system development. We reveal a negative correlation between the effects of immune cell traits on lifespan and COVID-19 risk. We find that lower B-cell CD19 levels are indicative of an increased risk of COVID-19 and decreased life expectancy, which is further validated by COVID-19 clinical data. Conclusions Our analysis suggests that the factors that accelerate aging lead to an increased COVID-19 risk and point to the importance of Notch signaling and B cells in both. Interventions that target these factors to reduce biological age may reduce the risk of COVID-19.
Collapse
Affiliation(s)
- Kejun Ying
- Biostatistics Group, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
- T. H. Chan School of Public Health, Harvard University, Boston, MA USA
| | - Ranran Zhai
- Biostatistics Group, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | | | - Anastasia V. Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Marco Mariotti
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Catalonia Spain
| | - Peter O. Fedichev
- Gero LLC PTE, Singapore City, Singapore
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region Russia
| | - Xia Shen
- Biostatistics Group, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
36
|
Ra R, Kim JS, Jeong KH, Hwang HS. COVID-19 and Sirolimus Treatment in a Kidney Transplant Recipient. EXP CLIN TRANSPLANT 2021; 19:977-980. [PMID: 34545780 DOI: 10.6002/ect.2021.0232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
One year after COVID-19 was declared a pandemic, the management of the disease in kidney transplant patients remains uncertain. The interruption of immunosuppressive therapy is frequently suggested in kidney transplant recipients with COVID-19; however, such an interruption potentially increases the risk of allograft rejection and hyperimmune response. We here report the successful treatment of COVID-19 pneumonia in a kidney transplant recipient who received a sirolimus-based regimen. The course of COVID-19 management was favorable for maintaining sirolimus treatment. Nevertheless, the patient showed signs of extreme overexposure to sirolimus because of drug interaction with antiviral treatment. This case illustrates the advantages and related adverse events of sirolimus-based immunosuppression in the management of COVID-19 in kidney transplant recipients.
Collapse
Affiliation(s)
- Ri Ra
- From the Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
37
|
Granata S, Carratù P, Stallone G, Zaza G. mTOR-Inhibition and COVID-19 in Kidney Transplant Recipients: Focus on Pulmonary Fibrosis. Front Pharmacol 2021; 12:710543. [PMID: 34497515 PMCID: PMC8419255 DOI: 10.3389/fphar.2021.710543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/11/2021] [Indexed: 12/24/2022] Open
Abstract
Kidney transplant recipients are at high risk of developing severe COVID-19 due to the coexistence of several transplant-related comorbidities (e.g., cardiovascular disease, diabetes) and chronic immunosuppression. As a consequence, a large part of SARS-CoV-2 infected patients have been managed with a reduction of immunosuppression. The mTOR-I, together with antimetabolites, have been often discontinued in order to minimize the risk of pulmonary toxicity and to antagonize pharmacological interaction with antiviral/anti-inflammatory drugs. However, at our opinion, this therapeutic strategy, although justified in kidney transplant recipients with severe COVID-19, should be carefully evaluated in asymptomatic/paucisymptomatic patients in order to avoid the onset of acute allograft rejections, to potentially exploit the mTOR-I antiviral properties, to reduce proliferation of conventional T lymphocytes (which could mitigate the cytokine storm) and to preserve Treg growth/activity which could reduce the risk of progression to severe disease. In this review, we discuss the current literature regarding the therapeutic potential of mTOR-Is in kidney transplant recipients with COVID-19 with a focus on pulmonary fibrosis.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Pierluigi Carratù
- Division of Internal Medicine, Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, "Aldo Moro" University of Bari, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| |
Collapse
|
38
|
Schoeman D, Fielding BC. Human Coronaviruses: Counteracting the Damage by Storm. Viruses 2021; 13:1457. [PMID: 34452323 PMCID: PMC8402835 DOI: 10.3390/v13081457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past 18 years, three highly pathogenic human (h) coronaviruses (CoVs) have caused severe outbreaks, the most recent causative agent, SARS-CoV-2, being the first to cause a pandemic. Although much progress has been made since the COVID-19 pandemic started, much about SARS-CoV-2 and its disease, COVID-19, is still poorly understood. The highly pathogenic hCoVs differ in some respects, but also share some similarities in clinical presentation, the risk factors associated with severe disease, and the characteristic immunopathology associated with the progression to severe disease. This review aims to highlight these overlapping aspects of the highly pathogenic hCoVs-SARS-CoV, MERS-CoV, and SARS-CoV-2-briefly discussing the importance of an appropriately regulated immune response; how the immune response to these highly pathogenic hCoVs might be dysregulated through interferon (IFN) inhibition, antibody-dependent enhancement (ADE), and long non-coding RNA (lncRNA); and how these could link to the ensuing cytokine storm. The treatment approaches to highly pathogenic hCoV infections are discussed and it is suggested that a greater focus be placed on T-cell vaccines that elicit a cell-mediated immune response, using rapamycin as a potential agent to improve vaccine responses in the elderly and obese, and the potential of stapled peptides as antiviral agents.
Collapse
Affiliation(s)
| | - Burtram C. Fielding
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa;
| |
Collapse
|
39
|
Kumari M, Kumar A. Can pharmaceutical drugs used to treat Covid-19 infection leads to human health risk? A hypothetical study to identify potential risk. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 778:146303. [PMID: 34030377 PMCID: PMC7942154 DOI: 10.1016/j.scitotenv.2021.146303] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 05/21/2023]
Abstract
This is the first study to assess human health risks due to the exposure of 'repurposed' pharmaceutical drugs used to treat Covid-19 infection. The study used a six-step approach to determine health risk estimates. For this, consumption of pharmaceuticals under normal circumstances and in Covid-19 infection was compiled to calculate the predicted environmental concentrations (PECs) in river water and in fishes. Risk estimates of pharmaceutical drugs were evaluated for adults as they are most affected by Covid-19 pandemic. Acceptable daily intakes (ADIs) are estimated using the no-observed-adverse-effect-level (NOAEL) or no observable effect level (NOEL) values in rats. The estimated ADI values are then used to calculate predicted no-effect concentrations (PNECs) for three different exposure routes (i) through the accidental ingestion of contaminated surface water during recreational activities only, (ii) through fish consumption only, and (iii) through combined accidental ingestion of contaminated surface water during recreational activities and fish consumption. Higher risk values (hazard quotient, HQ: 337.68, maximum; 11.83, minimum) were obtained for the combined ingestion of contaminated water during recreational activities and fish consumption exposure under the assumptions used in this study indicating possible effects to human health. Amongst the pharmaceutical drugs, ritonavir emerged as main drug, and is expected to pose adverse effects on r human health through fish consumption. Mixture toxicity analysis showed major risk effects of exposure of pharmaceutical drugs (interaction-based hazard index, HIint: from 295.42 (for lopinavir + ritonavir) to 1.20 for chloroquine + rapamycin) demonstrating possible risks due to the co-existence of pharmaceutical in water. The presence of background contaminants in contaminated water does not show any influence on the observed risk estimates as indicated by low HQadd values (<1). Regular monitoring of pharmaceutical drugs in aquatic environment needs to be carried out to reduce the adverse effects of pharmaceutical drugs on human health.
Collapse
Affiliation(s)
- Minashree Kumari
- Department of Civil Engineering, Indian Institute of Technology Delhi, Hauz Khas, Delhi 110016, India.
| | - Arun Kumar
- Department of Civil Engineering, Indian Institute of Technology Delhi, Hauz Khas, Delhi 110016, India.
| |
Collapse
|
40
|
Zhang S, Asquith B, Szydlo R, Tregoning JS, Pollock KM. Peripheral T cell lymphopenia in COVID-19: potential mechanisms and impact. IMMUNOTHERAPY ADVANCES 2021; 1:ltab015. [PMID: 35965490 PMCID: PMC9364037 DOI: 10.1093/immadv/ltab015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/10/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022] Open
Abstract
Immunopathogenesis involving T lymphocytes, which play a key role in defence against viral infection, could contribute to the spectrum of COVID-19 disease and provide an avenue for treatment. To address this question, a review of clinical observational studies and autopsy data in English and Chinese languages was conducted with a search of registered clinical trials. Peripheral lymphopenia affecting CD4 and CD8 T cells was a striking feature of severe COVID-19 compared with non-severe disease. Autopsy data demonstrated infiltration of T cells into organs, particularly the lung. Seventy-four clinical trials are on-going that could target T cell-related pathogenesis, particularly IL-6 pathways. SARS-CoV-2 infection interrupts T cell circulation in patients with severe COVID-19. This could be due to redistribution of T cells into infected organs, activation induced exhaustion, apoptosis, or pyroptosis. Measuring T cell dynamics during COVID-19 will inform clinical risk-stratification of hospitalised patients and could identify those who would benefit most from treatments that target T cells.
Collapse
Affiliation(s)
- Sifan Zhang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Richard Szydlo
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Katrina M Pollock
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
41
|
Mashayekhi-Sardoo H, Hosseinjani H. A new application of mTOR inhibitor drugs as potential therapeutic agents for COVID-19. J Basic Clin Physiol Pharmacol 2021; 33:17-25. [PMID: 34218545 DOI: 10.1515/jbcpp-2020-0495] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
Since December 2019, the COVID-19 emerging pandemic caused by SARS-CoV-2 has resulted in one of the most important global health threats. Concerning the absence of an approved effective vaccine or drug for the treatment and outcome improvement of COVID-19 patients, and the role of SARS-CoV-2 in activation of mammalian target of rapamycin (mTOR) pathway, we decided to review the previous data regarding the therapeutic effect of mTOR inhibitor drugs in COVID-19 patients. We searched the scientific databases such as Web of Science, Embase, Medline (PubMed), Scopus, and Google Scholar using appropriate keywords to find suitable studies or suggestions until October 2020. The findings of the current study confirmed that mTOR inhibitor drugs through suggested mechanisms such as T cell adjustment, induction of autophagy without apoptosis, reduction of viral replication, restoration of the T-cell function, decrease cytokine storm, and moderation of the mTOR-PI3K-AKT pathway activation bring about a therapeutic impact in COVID-19 patients. Taken together, it is necessary to find a suitable therapy for the COVID-19 pandemic emerging. In this regard, we clarify that it is valuable to consider the therapeutic effect of mTOR inhibitor drugs and metformin by its mTOR inhibition property in the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesamoddin Hosseinjani
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
42
|
van Eijk LE, Binkhorst M, Bourgonje AR, Offringa AK, Mulder DJ, Bos EM, Kolundzic N, Abdulle AE, van der Voort PHJ, Olde Rikkert MGM, van der Hoeven JG, den Dunnen WFA, Hillebrands J, van Goor H. COVID-19: immunopathology, pathophysiological mechanisms, and treatment options. J Pathol 2021; 254:307-331. [PMID: 33586189 PMCID: PMC8013908 DOI: 10.1002/path.5642] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to spread globally despite the worldwide implementation of preventive measures to combat the disease. Although most COVID-19 cases are characterised by a mild, self-limiting disease course, a considerable subset of patients develop a more severe condition, varying from pneumonia and acute respiratory distress syndrome (ARDS) to multi-organ failure (MOF). Progression of COVID-19 is thought to occur as a result of a complex interplay between multiple pathophysiological mechanisms, all of which may orchestrate SARS-CoV-2 infection and contribute to organ-specific tissue damage. In this respect, dissecting currently available knowledge of COVID-19 immunopathogenesis is crucially important, not only to improve our understanding of its pathophysiology but also to fuel the rationale of both novel and repurposed treatment modalities. Various immune-mediated pathways during SARS-CoV-2 infection are relevant in this context, which relate to innate immunity, adaptive immunity, and autoimmunity. Pathological findings in tissue specimens of patients with COVID-19 provide valuable information with regard to our understanding of pathophysiology as well as the development of evidence-based treatment regimens. This review provides an updated overview of the main pathological changes observed in COVID-19 within the most commonly affected organ systems, with special emphasis on immunopathology. Current management strategies for COVID-19 include supportive care and the use of repurposed or symptomatic drugs, such as dexamethasone, remdesivir, and anticoagulants. Ultimately, prevention is key to combat COVID-19, and this requires appropriate measures to attenuate its spread and, above all, the development and implementation of effective vaccines. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Larissa E van Eijk
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Mathijs Binkhorst
- Department of Paediatrics, Subdivision of NeonatologyRadboud University Medical Center Amalia Children's HospitalNijmegenThe Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Annette K Offringa
- Microbiology and System BiologyNetherlands Organisation for Applied Scientific ResearchZeistThe Netherlands
| | - Douwe J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Eelke M Bos
- Department of NeurosurgeryErasmus University Medical CenterRotterdamThe Netherlands
| | - Nikola Kolundzic
- Stem Cell Laboratory, Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Assisted Conception Unit, Guy's HospitalLondonUK
| | - Amaal E Abdulle
- Department of Internal Medicine, Division of Vascular Medicine, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Peter HJ van der Voort
- Department of Critical Care, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Marcel GM Olde Rikkert
- Department of Geriatric MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Wilfred FA den Dunnen
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Jan‐Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
43
|
Mohiuddin M, Kasahara K. Cellular senescence is a potential severity factor for COVID-19: Suitable targets required to eliminate cellular senescence. Respir Med 2021; 186:106517. [PMID: 34186259 PMCID: PMC8215876 DOI: 10.1016/j.rmed.2021.106517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/18/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Md Mohiuddin
- Department of Respiratory Medicine, Kanazawa University, Ishikawa, Japan.
| | - Kazuo Kasahara
- Department of Respiratory Medicine, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
44
|
Lange A, Lange J, Jaskuła E. Cytokine Overproduction and Immune System Dysregulation in alloHSCT and COVID-19 Patients. Front Immunol 2021; 12:658896. [PMID: 34149697 PMCID: PMC8206782 DOI: 10.3389/fimmu.2021.658896] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
The COVID-19 pathomechanism depends on (i) the pathogenicity of the virus, (ii) ability of the immune system to respond to the cytopathic effect of the virus infection, (iii) co-morbidities. Inflammatory cytokine production constitutes a hallmark of COVID-19 that is facilitated by inability of adaptive immunity to control virus invasion. The effect of cytokine release syndrome is deleterious, but the severity of it depends on other confounding factors: age and comorbidities. In this study, we analyze the literature data on the post-transplant course of allogeneic hematopoietic stem cell transplanted (alloHSCT) patients, which is affected by generated inflammatory cytokines. The sequence of events boosting cytokine production was analyzed in relation to clinical and laboratory data highlighting the impact of cytokine generation on the post-transplant course. The collected data were compared to those from studies on COVID-19 patients. The similarities are: (i) the damage/pathogen-associated molecular pattern (DAMP/PAMP) stage is similar except for the initiation hit being sterile in alloHSCT (toxic damage of conditioning regimen) and viral in COVID-19; (ii) genetic host-derived factors play a role; (iii) adaptive immunity fails, DAMP signal(s) increases, over-production of cytokines occurs; (iv) monocytes lacking HLADR expression emerge, being suppressor cells hampering adaptive immunity; (v) immune system homeostasis is broken, the patient's status deteriorates to bed dependency, leading to hypo-oxygenation and malnutrition, which in turn stimulates the intracellular alert pathways with vigorous transcription of cytokine genes. All starts with the interaction between DAMPs with appropriate receptors, which leads to the production of pro-inflammatory cytokines, the inflammatory process spreads, tissue is damaged, DAMPs are released and a vicious cycle occurs. Attempts to modify intracellular signaling pathways in patients with post-alloHSCT graft vs host disease have already been undertaken. The similarities documented in this study show that this approach may also be used in COVID-19 patients for tuning signal transduction processes to interrupt the cycle that powers the cytokine overproduction.
Collapse
Affiliation(s)
- Andrzej Lange
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| | - Janusz Lange
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| | - Emilia Jaskuła
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Lower Silesian Center for Cellular Transplantation with National Bone Marrow Donor Registry, Wroclaw, Poland
| |
Collapse
|
45
|
Mohiuddin M, Kasahara K. The emerging role of cellular senescence in complications of COVID-19. Cancer Treat Res Commun 2021; 28:100399. [PMID: 34023769 PMCID: PMC8123375 DOI: 10.1016/j.ctarc.2021.100399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has triggered a sudden global change in healthcare systems. Cancer patients have a higher risk of death from COVID-19 in comparison to patients without cancer. Many studies have stated that various factors, such as older age, frequent exposure to healthcare, and higher smoking rates are responsible for the complications of COVID-19. We hypothesize that side effects of chemotherapy, such as cellular senescence, could worsen COVID-19. Given this situation, in this review, we highlight the updated findings of research investigating the impact of cellular senescence on COVID-19 complications and explored potential therapeutic targets for eliminating senescent cells during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Md Mohiuddin
- Department of Respiratory Medicine, Kanazawa University, Kanazawa 920-8641, Ishikawa, Japan.
| | - Kazuo Kasahara
- Department of Respiratory Medicine, Kanazawa University, Kanazawa 920-8641, Ishikawa, Japan
| |
Collapse
|
46
|
Pereira GJDS, Leão AHFF, Erustes AG, Morais IBDM, Vrechi TADM, Zamarioli LDS, Pereira CAS, Marchioro LDO, Sperandio LP, Lins ÍVF, Piacentini M, Fimia GM, Reckziegel P, Smaili SS, Bincoletto C. Pharmacological Modulators of Autophagy as a Potential Strategy for the Treatment of COVID-19. Int J Mol Sci 2021; 22:4067. [PMID: 33920748 PMCID: PMC8071111 DOI: 10.3390/ijms22084067] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
The family of coronaviruses (CoVs) uses the autophagy machinery of host cells to promote their growth and replication; thus, this process stands out as a potential target to combat COVID-19. Considering the different roles of autophagy during viral infection, including SARS-CoV-2 infection, in this review, we discuss several clinically used drugs that have effects at different stages of autophagy. Among them, we mention (1) lysosomotropic agents, which can prevent CoVs infection by alkalinizing the acid pH in the endolysosomal system, such as chloroquine and hydroxychloroquine, azithromycin, artemisinins, two-pore channel modulators and imatinib; (2) protease inhibitors that can inhibit the proteolytic cleavage of the spike CoVs protein, which is necessary for viral entry into host cells, such as camostat mesylate, lopinavir, umifenovir and teicoplanin and (3) modulators of PI3K/AKT/mTOR signaling pathways, such as rapamycin, heparin, glucocorticoids, angiotensin-converting enzyme inhibitors (IECAs) and cannabidiol. Thus, this review aims to highlight and discuss autophagy-related drugs for COVID-19, from in vitro to in vivo studies. We identified specific compounds that may modulate autophagy and exhibit antiviral properties. We hope that research initiatives and efforts will identify novel or "off-label" drugs that can be used to effectively treat patients infected with SARS-CoV-2, reducing the risk of mortality.
Collapse
Affiliation(s)
- Gustavo José da Silva Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Anderson Henrique França Figueredo Leão
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Ingrid Beatriz de Melo Morais
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Talita Aparecida de Moraes Vrechi
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Lucas dos Santos Zamarioli
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Cássia Arruda Souza Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Laís de Oliveira Marchioro
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Letícia Paulino Sperandio
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Ísis Valeska Freire Lins
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘La Zaro Spallanzani’, 00149 Rome, Italy;
| | - Gian Maria Fimia
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘La Zaro Spallanzani’, 00149 Rome, Italy;
- Department of Molecular Medicine, University of Rome La Sapienza, 00185 Rome, Italy
| | - Patrícia Reckziegel
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Claudia Bincoletto
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| |
Collapse
|
47
|
Adenote A, Dumic I, Madrid C, Barusya C, Nordstrom CW, Rueda Prada L. NAFLD and Infection, a Nuanced Relationship. Can J Gastroenterol Hepatol 2021; 2021:5556354. [PMID: 33977096 PMCID: PMC8087474 DOI: 10.1155/2021/5556354] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has increased significantly over the last few decades mirroring the increase in obesity and type II diabetes mellitus. NAFLD has become one of the most common indications for liver transplantation. The deleterious effects of NAFLD are not isolated to the liver only, for it has been recognized as a systemic disease affecting multiple organs through protracted low-grade inflammation mediated by the metabolic activity of excessive fat tissue. Extrahepatic manifestations of NAFLD such as cardiovascular disease, polycystic ovarian syndrome, chronic kidney disease, and hypothyroidism have been well described in the literature. In recent years, it has become evident that patients suffering from NAFLD might be at higher risk of developing various infections. The proposed mechanism for this association includes links through hyperglycemia, insulin resistance, alterations in innate immunity, obesity, and vitamin D deficiency. Additionally, a risk independent of these factors mediated by alterations in gut microbiota might contribute to a higher burden of infections in these individuals. In this narrative review, we synthetize current knowledge on several infections including urinary tract infection, pneumonia, Helicobacter pylori, coronavirus disease 2019, and Clostridioides difficile as they relate to NAFLD. Additionally, we explore NAFLD's association with hidradenitis suppurativa.
Collapse
Affiliation(s)
- Abimbola Adenote
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Igor Dumic
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Cristian Madrid
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Christopher Barusya
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Charles W. Nordstrom
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Libardo Rueda Prada
- Division of Hospital Internal Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
48
|
Stoy N. Involvement of Interleukin-1 Receptor-Associated Kinase 4 and Interferon Regulatory Factor 5 in the Immunopathogenesis of SARS-CoV-2 Infection: Implications for the Treatment of COVID-19. Front Immunol 2021; 12:638446. [PMID: 33936053 PMCID: PMC8085890 DOI: 10.3389/fimmu.2021.638446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) and interferon regulatory factor 5 (IRF5) lie sequentially on a signaling pathway activated by ligands of the IL-1 receptor and/or multiple TLRs located either on plasma or endosomal membranes. Activated IRF5, in conjunction with other synergistic transcription factors, notably NF-κB, is crucially required for the production of proinflammatory cytokines in the innate immune response to microbial infection. The IRAK4-IRF5 axis could therefore have a major role in the induction of the signature cytokines and chemokines of the hyperinflammatory state associated with severe morbidity and mortality in COVID-19. Here a case is made for considering IRAK4 or IRF5 inhibitors as potential therapies for the "cytokine storm" of COVID-19.
Collapse
Affiliation(s)
- Nicholas Stoy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Pal N, Mavi AK, Kumar S, Kumar U, Joshi MD, Saluja R. Current updates on adaptive immune response by B cell and T cell stimulation and therapeutic strategies for novel coronavirus disease 2019 (COVID-19) treatment. Heliyon 2021; 7:e06894. [PMID: 33937545 PMCID: PMC8076978 DOI: 10.1016/j.heliyon.2021.e06894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/09/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
The prevalence of COVID-19 continues to rise with more than 114,315,846 million confirmed cases and 2,539,427 deaths worldwide by 3 March 2021 and this number kept on increasing day by day. There is no clear therapeutic treatment or vaccine available for COVID-19 till date and by seeing such a high rise in the cases of COVID-19 on daily basis, it would have been necessary to implement precautions and hygienic measures to monitor and reduce human-to-human transmission of SARS-CoV-2 before there is any successful intervention/treatment available. Currently, several studies demonstrated the important improvements in both the innate and adaptive immune systems of COVID-19 patients. In particular, pre-existing research, on immune response to B cell and T cells are highlighting that pre-existing immunity exists in about 90% of the general population because of previous exposure to CoVs and having immunity against these CoVs. Although it is not clear from, the current studies on COVID-19 but it assumed that, it might have implication to COVID-19 severity and could play an important role in treatment or vaccine development against COVID-19. This review summarizes the information from occurrence of SARS-CoV-2 to its pathogenesis, transmission, adaptive immune response with respect to T cell and B cell stimulation and therapeutic interventions/treatment against COVID-19.
Collapse
Affiliation(s)
- Neeraj Pal
- Department of Molecular Biology and Genetic Engineering, GB Pant University of Agriculture and Technology, Pantnagar, 263145, India
| | - Anil Kumar Mavi
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, 110007, India
| | - Sundip Kumar
- Department of Molecular Biology and Genetic Engineering, GB Pant University of Agriculture and Technology, Pantnagar, 263145, India
| | - Umesh Kumar
- School of Biosciences, IMS Ghaziabad University Courses Campus, Uttar Pradesh, 201015, India
| | - Maya Datt Joshi
- Department of Biotechnology, Shobhit Institute of Engineering & Technology (Deemed to be University), Meerut, 250110, India
| | - Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| |
Collapse
|
50
|
Harwansh RK, Bahadur S. Herbal Medicine in Fighting Against COVID-19: New Battle with an Old Weapon. Curr Pharm Biotechnol 2021; 23:235-260. [PMID: 33749558 DOI: 10.2174/1389201022666210322124348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 11/22/2022]
Abstract
World population has been suffering due to the outbreak of present pandemic situation of COVID-19. The disease has become life-threatening in a very short time with touching on most of the citizenry and economic systems globally. The novel virus, SARS-CoV-2 has been known as the causative agent of COVID-19. The SARS-CoV-2 is single stranded RNA virus having ~30 kb genomic components which are 70% identical to SARS-CoV. The main process of pathophysiology of COVID-19 has been associated with the interaction of a novel coronavirus with host cell receptor, angiotensin-converting enzyme-2 (ACE 2) by fusion. Therapeutic agents having serine protease inhibitors and ACE-2 blockers may be explored for the treatment by inhibiting the viral target such as Mpro, RdRp, PLpro and helicase. Herbal medicine has a wide array chemical entity with potential health benefits including antiviral activity which may be explored as alternative treatment of COVID-19. The herbal bioactives like catechins, andrographolide, hesperidin, biorobin, scutellarein, silvestrol, shikonin, tryptanthrin, vitexin quercetin, myricetin, caffeic acid, psoralidin, luteolin etc have showed potential inhibitory effect against SARS-CoV-2. Recent research reports indicate that the various plant secondary metabolites have shown the potential antiviral activities. The present review article highlights on the recent information on the mechanism of actions and applications of herbal medicine in the treatment of COVID-19.
Collapse
Affiliation(s)
- Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura - 281406. India
| | - Shiv Bahadur
- Institute of Pharmaceutical Research, GLA University, Mathura - 281406. India
| |
Collapse
|