1
|
Lee E, Yang D, Hong JH. Prominent Naturally Derived Oxidative-Stress-Targeting Drugs and Their Applications in Cancer Treatment. Antioxidants (Basel) 2025; 14:49. [PMID: 39857383 PMCID: PMC11760868 DOI: 10.3390/antiox14010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
The relationship between oxidative stress and cancer has been extensively studied and highlighted, along with its role in various aspects of angiogenesis. The modulation of oxidative levels and the adaptive mechanisms of oxidative stress in cancer systems are attractive research themes for developing anti-cancer strategies. Reactive oxygen species (ROS) are involved in various pathophysiological processes and play crucial roles in DNA damage and angiogenesis. Although cancer cells have developed various adaptive defense mechanisms against oxidative stress, excessive ROS production has been proposed as an anti-cancer strategy to induce cellular apoptosis. In particular, natural-source-based antioxidants have been identified as effective against cancers, and various delivery platforms have been developed to enhance their efficacy. In this review, we highlighted the anti-cancer components (plumbagin, quercetin, resveratrol, curcumin, xanthatin, carvacrol, telmisartan, and sulforaphane) that modulate ROS levels and the recent targeting platforms used to increase the application of anti-cancer drugs and the developed delivery platforms with diverse mechanisms of action. Further, we summarized the actual doses used and the effects of these drug candidates in various cancer systems. Overall, this review provides beneficial research themes for expanding cancer-targeting fields and addressing limited applications in diverse cancer types.
Collapse
Affiliation(s)
| | - Dongki Yang
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| | - Jeong Hee Hong
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
2
|
Bermúdez-Abreut E, Fundora-Barrios T, Hernández Fernández DR, Noa Romero E, Fraga-Quintero A, Casadesús Pazos AV, Fernández-Marrero B, Plasencia Iglesias CA, Clavel Pérez M, Sosa Aguiar K, Sánchez-Ramírez B, Hernández T. Antiviral activity of an ACE2-Fc fusion protein against SARS-CoV-2 and its variants. PLoS One 2025; 20:e0312402. [PMID: 39752453 PMCID: PMC11698409 DOI: 10.1371/journal.pone.0312402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 10/04/2024] [Indexed: 01/06/2025] Open
Abstract
SARS-CoV-2 has continued spreading around the world in recent years since the initial outbreak in 2019, frequently developing into new variants with greater human infectious capacity. SARS-CoV-2 and its mutants use the angiotensin-converting enzyme 2 (ACE2) as a cellular entry receptor, which has triggered several therapeutic strategies against COVID-19 relying on the use of ACE2 recombinant proteins as decoy receptors. In this work, we propose an ACE2 silent Fc fusion protein (ACE2-hFcLALA) as a candidate therapy against COVID-19. This fusion protein was able to block the binding of SARS-CoV-2 RBD to ACE2 receptor as measured by ELISA and flow cytometry inhibition assays. Moreover, we used classical neutralization assays and a progeny neutralization assay to show that the ACE2-hFcLALA fusion protein is capable of neutralizing the authentic virus. Additionally, we found that this fusion protein was more effective in preventing in vitro infection with different variants of interest (alpha, beta, delta, and omicron) compared to the D614G strain. Our results suggest the potential of this molecule to be used in both therapeutic and preventive settings against current and emerging mutants that use ACE2 as a gateway to human cells.
Collapse
Affiliation(s)
| | - Talia Fundora-Barrios
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Enrique Noa Romero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Anitza Fraga-Quintero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Ana V. Casadesús Pazos
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | | | - Marilyn Clavel Pérez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Katya Sosa Aguiar
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Tays Hernández
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| |
Collapse
|
3
|
Nisar H, Wajid B, Anwar F, Ahmad A, Javaid A, Attique SA, Nisar W, Saeed A, Shahid S, Sadaf S. Bioinformatics and systems biology analysis revealed PMID26394986-Compound-10 as potential repurposable drug against covid-19. J Biomol Struct Dyn 2024; 42:7972-7985. [PMID: 37534820 DOI: 10.1080/07391102.2023.2242500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
The global health pandemic known as COVID-19, which stems from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a significant concern worldwide. Several treatment methods exist for COVID-19; however, there is an urgent demand for previously established drugs and vaccines to effectively combat the disease. Since, discovering new drugs poses a significant challenge, making the repurposing of existing drugs can potentially reduce time and costs compared to developing entirely new drugs from scratch. The objective of this study is to identify hub genes and associated repurposed drugs targeting them. We analyzed differentially expressed genes (DEGs) by analyzing RNA-seq transcriptomic datasets and integrated with genes associated with COVID-19 present in different databases. We detected 173 Covid-19 associated genes for the construction of a protein-protein interaction (PPI) network which resulted in the identification of the top 10 hub genes/proteins (STAT1, IRF7, MX1, IRF9, ISG15, OAS3, OAS2, OAS1, IRF3, and IRF1). Hub genes were subjected to GO functional and KEGG pathway enrichment analyses, which indicated some key roles and signaling pathways that were strongly related to SARS-CoV-2 infections. We conducted drug repurposing analysis using CMap, TTD, and DrugBank databases with these 10 hub genes, leading to the identification of Piceatannol, CKD-712, and PMID26394986-Compound-10 as top-ranked candidate drugs. Finally, drug-gene interactions analysis through molecular docking and validated via molecular dynamic simulation for 80 ns suggests PMID26394986-Compound-10 as the only potential drug. Our research demonstrates how in silico analysis might produce repurposing candidates to help respond faster to new disease outbreaks.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Haseeb Nisar
- Department of Life-Sciences, University of Management and Technology, Lahore, Pakistan
| | - Bilal Wajid
- Ibn Sina Research & Development Division, Sabz-Qalam, Lahore, Pakistan
- Department of Electrical Engineering, University of Engineering and Technology, Lahore, Pakistan
| | | | - Ashfaq Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Anum Javaid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Syed Awais Attique
- Bioinformatics Institute, Agency for Science, Technology and Research (A(*)STAR), Singapore, Singapore
| | - Wardah Nisar
- Department of Public Health, University of Health Sciences, Lahore, Pakistan
| | - Amir Saeed
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Samiah Shahid
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
4
|
Samy W, Gaber OA, Amer RM, El-Deeb NA, Abdelmoaty AA, Sharaf AL, El-Gebaly AM, Mosbah R, Alsadik ME, Fawzy A, Ahmed AA. Role of ACE2 and TMPRSS2 polymorphisms in clinical severity and outcomes of COVID-19 in Egypt. Afr J Lab Med 2024; 13:2375. [PMID: 39228902 PMCID: PMC11369560 DOI: 10.4102/ajlm.v13i1.2375] [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: 11/21/2023] [Accepted: 05/23/2024] [Indexed: 09/05/2024] Open
Abstract
Background The clinical presentations of coronavirus disease 2019 (COVID-19) exhibit significant variation, ranging from asymptomatic cases to mortality resulting from severe pneumonia. Host genetics can partially explain this variation. Objective This study evaluated possible associations between severity and outcome of COVID-19 and single nucleotide polymorphism (SNP) rs2285666 in the ACE2 gene and SNP rs2070788 in the TMPRSS2 gene. Methods The study included a sample of 100 consecutive adult patients admitted to the COVID-19 Isolation and Intensive Care Units of the Zagazig University Hospitals, Zagazig, Egypt from July 2021 to November 2021. For rs2285666, polymerase chain reaction-restriction fragment length polymorphism was carried out. For rs2070788, real-time polymerase chain reaction was performed. Results For rs2285666, the GA genotype was the most frequent among female patients (39% [16/41]) and the A genotype was more prevalent among male patients (54.2% [32/59]). For rs2070788, the AA genotype was the most frequent among all patients (46% [46/100]). No rs2285666 or rs2070788 genotypes or allele frequencies had significant associations with either severity or outcomes of patients. Conclusion This study found no significant associations of COVID-19 severity or outcomes of patients with genotypes or allele frequencies of the rs2285666 SNP in the ACE2 gene or the rs2070788 SNP of the TMPRSS2 gene. The search for other genetic associations with COVID-19 infection is still required. What this study adds The study reveals that host genetics explain the variation observed in the disease. Specific genetic variants can confer either increased susceptibility or resistance to the disease.
Collapse
Affiliation(s)
- Walaa Samy
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Osama A Gaber
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rania M Amer
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nahawand A El-Deeb
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed A Abdelmoaty
- Department of Tropical Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed L Sharaf
- Department of Tropical Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed M El-Gebaly
- Department of Tropical Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rasha Mosbah
- Department of Infection Prevention and Control, Zagazig University Hospitals, Zagazig University, Zagazig, Egypt
| | - Maha E Alsadik
- Chest Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amal Fawzy
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Alshymaa A Ahmed
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
5
|
Pauly I, Kumar Singh A, Kumar A, Singh Y, Thareja S, Kamal MA, Verma A, Kumar P. Current Insights and Molecular Docking Studies of the Drugs under Clinical Trial as RdRp Inhibitors in COVID-19 Treatment. Curr Pharm Des 2023; 28:3677-3705. [PMID: 36345244 DOI: 10.2174/1381612829666221107123841] [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: 05/27/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022]
Abstract
Study Background & Objective: After the influenza pandemic (1918), COVID-19 was declared a Vth pandemic by the WHO in 2020. SARS-CoV-2 is an RNA-enveloped single-stranded virus. Based on the structure and life cycle, Protease (3CLpro), RdRp, ACE2, IL-6, and TMPRSS2 are the major targets for drug development against COVID-19. Pre-existing several drugs (FDA-approved) are used to inhibit the above targets in different diseases. In coronavirus treatment, these drugs are also in different clinical trial stages. Remdesivir (RdRp inhibitor) is the only FDA-approved medicine for coronavirus treatment. In the present study, by using the drug repurposing strategy, 70 preexisting clinical or under clinical trial molecules were used in scrutiny for RdRp inhibitor potent molecules in coronavirus treatment being surveyed via docking studies. Molecular simulation studies further confirmed the binding mechanism and stability of the most potent compounds. MATERIAL AND METHODS Docking studies were performed using the Maestro 12.9 module of Schrodinger software over 70 molecules with RdRp as the target and remdesivir as the standard drug and further confirmed by simulation studies. RESULTS The docking studies showed that many HIV protease inhibitors demonstrated remarkable binding interactions with the target RdRp. Protease inhibitors such as lopinavir and ritonavir are effective. Along with these, AT-527, ledipasvir, bicalutamide, and cobicistat showed improved docking scores. RMSD and RMSF were further analyzed for potent ledipasvir and ritonavir by simulation studies and were identified as potential candidates for corona disease. CONCLUSION The drug repurposing approach provides a new avenue in COVID-19 treatment.
Collapse
Affiliation(s)
- Irine Pauly
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jaddah, Saudi Arabia.,Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia.,Novel Global Community Educational Foundation, Australia Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, Australia
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, 151401, India
| |
Collapse
|
6
|
Banik SP, Bhattacharyya M, Ghosh R, Chatterjee T, Basak P. Unveiling the prevalence and impact of diabetes on COVID-19. VIRAL, PARASITIC, BACTERIAL, AND FUNGAL INFECTIONS 2023:287-301. [DOI: 10.1016/b978-0-323-85730-7.00045-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Verma R, Raj S, Berry U, Ranjith-Kumar CT, Surjit M. Drug Repurposing for COVID-19 Therapy: Pipeline, Current Status and Challenges. DRUG REPURPOSING FOR EMERGING INFECTIOUS DISEASES AND CANCER 2023:451-478. [DOI: 10.1007/978-981-19-5399-6_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
El Ouafi Z, Rhalem W, Habib N, Idrissi Azami A, Sehli S, Al Idrissi N, Bakkali F, Abderrazak R, Merzouki M, Allali I, Amzazi S, Nejjari C, Ghazal H. Molecular Modeling Targeting the ACE2 Receptor with Cannabis sativa's Active Ingredients for Antiviral Drug Discovery against SARS-CoV-2 Infections. Bioinform Biol Insights 2022; 16:11779322221145380. [PMID: 36582392 PMCID: PMC9793058 DOI: 10.1177/11779322221145380] [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: 08/31/2022] [Accepted: 11/25/2022] [Indexed: 12/25/2022] Open
Abstract
The emergence of a novel coronavirus that later on rendered a global pandemic, caused desperation within the communities and drove increased interest in exploring medicinal plant-based therapeutics to treat and prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infections. Many medicinal plants have been reported to have antiviral, anti-inflammatory, and immunomodulatory effects that hinder, cure, or ease the symptoms of COVID-19 infection. This exploratory study seeks to dock the active components of Cannabis sativa, a natural plant with several pharmacological and biological properties, with the angiotensin-converting enzyme II (ACE2) receptor. A total of 3 C. sativa active components have been found to bind to the ACE2 protein active site and could inhibit spike binding, although they do not compete directly with the receptor-binding domain (RBD) of SARS-CoV-2. 6-Prenylapigenin, cannabivarin (CBN-C3), and Δ8-tetrahydrocannabinolic acid-A (Δ8-THCA) have a greater affinity (-8.3, -8.3, and -8.0 kcal/mol, respectively) and satisfactory interaction with ACE2 than its inhibitor MLN-4760 (-7.1 kcal/mol). These potential drugs with higher affinity for the ACE2 receptor and adequate absorption, distribution, metabolism, excretion, and toxicity (ADMET) values are candidates for treating or preventing SARS-CoV-2 infections. In vitro and in vivo investigations are needed to evaluate further the efficacy and toxicity of these hit compounds.
Collapse
Affiliation(s)
- Zainab El Ouafi
- Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Casablanca, Morocco
| | - Wajih Rhalem
- Electronic Systems, Sensors and
Nanobiotechnologies (E2SN), École Nationale Supérieure des Arts et Métiers (ENSAM),
Mohammed V University, Rabat, Morocco
| | - Nihal Habib
- Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Casablanca, Morocco
| | - Abdellah Idrissi Azami
- Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Casablanca, Morocco
| | - Sofia Sehli
- Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Casablanca, Morocco
| | - Najib Al Idrissi
- Department of Surgery, School of
Medicine, Mohammed VI University of Health Sciences Casablanca, Casablanca,
Morocco
| | - Fadil Bakkali
- Toxicology Laboratory, School of
Medicine, Mohammed VI University of Health Sciences (UM6SS) Casablanca, Casablanca,
Morocco
| | - Rfaki Abderrazak
- National Center for Scientific and
Technical Research (CNRST), Rabat, Morocco
| | - Mohamed Merzouki
- Bioengineering Laboratory, Faculty of
Science and Technologies, University of Sultan Moulay Slimane, Beni Mellal,
Morocco
| | - Imane Allali
- Laboratory of Human Pathologies
Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human
Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat,
Rabat, Morocco
| | - Saaïd Amzazi
- Laboratory of Human Pathologies
Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human
Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat,
Rabat, Morocco
| | - Chakib Nejjari
- Department of Epidemiology and
Biostatistics, International School of Public Health, Mohammed VI University of
Health Sciences Casablanca, Casablanca, Morocco,Department of Epidemiology and Public
Health, Faculty of Medicine, University Sidi Mohammed Ben Abdellah, Fes,
Morocco
| | - Hassan Ghazal
- Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Casablanca, Morocco,Electronic Systems, Sensors and
Nanobiotechnologies (E2SN), École Nationale Supérieure des Arts et Métiers (ENSAM),
Mohammed V University, Rabat, Morocco,National Center for Scientific and
Technical Research (CNRST), Rabat, Morocco,Hassan Ghazal, Laboratory of Genomics and
Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences
Casablanca, Bld Mohammed Taieb Naciri, Hay Hassani, Casablanca, 82 403, Morocco.
| |
Collapse
|
9
|
Mitra D, Paul M, Thatoi H, Mohapatra PKD. Study of potentiality of dexamethasone and its derivatives against Covid-19. J Biomol Struct Dyn 2022; 40:10239-10249. [PMID: 34182880 DOI: 10.1080/07391102.2021.1942210] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In December 2019, COVID-19 epidemic was reported in Wuhan, China, and subsequently the infection has spread all over the world and became pandemic. The death toll associated with the pandemic is increasing day by day in a high rate. Herein, an effort has been made to identify the potentiality of commercially available drugs and also their probable derivatives for creation of better opportunity to make more powerful drugs against coronavirus. This study involves the in-silico interactions of dexamethasone and its derivatives against the multiple proteins of SARS-CoV-2 with the help of various computational methods. Descriptor parameters revealed their non-toxic effect in the human body. Ultimately docking studies and molecular dynamic simulation on those target protein by dexamethasone and its derivatives showed a high binding energy. Dexamethasone showed -9.8 kcal/mol and its derivative D5 showed -12.1 kcal/mol binding energy. Those scores indicate that dexamethasone has more therapeutic effect on SARS CoV-2 than other currently used drugs. Derivatives give the clue for the synthesis of a novel drug to remove SARS CoV-2. Until then, dexamethasone will be used as a potential inhibitor of SARS CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Debanjan Mitra
- Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| | - Manish Paul
- Department of Biotechnology, Maharaja Sriram Chandra Bhanja Deo University, Baripada, Odisha, India
| | - Hrudayanath Thatoi
- Department of Biotechnology, Maharaja Sriram Chandra Bhanja Deo University, Baripada, Odisha, India
| | | |
Collapse
|
10
|
Manan A, Pirzada RH, Haseeb M, Choi S. Toll-like Receptor Mediation in SARS-CoV-2: A Therapeutic Approach. Int J Mol Sci 2022; 23:10716. [PMID: 36142620 PMCID: PMC9502216 DOI: 10.3390/ijms231810716] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/10/2022] [Accepted: 09/10/2022] [Indexed: 01/18/2023] Open
Abstract
The innate immune system facilitates defense mechanisms against pathogen invasion and cell damage. Toll-like receptors (TLRs) assist in the activation of the innate immune system by binding to pathogenic ligands. This leads to the generation of intracellular signaling cascades including the biosynthesis of molecular mediators. TLRs on cell membranes are adept at recognizing viral components. Viruses can modulate the innate immune response with the help of proteins and RNAs that downregulate or upregulate the expression of various TLRs. In the case of COVID-19, molecular modulators such as type 1 interferons interfere with signaling pathways in the host cells, leading to an inflammatory response. Coronaviruses are responsible for an enhanced immune signature of inflammatory chemokines and cytokines. TLRs have been employed as therapeutic agents in viral infections as numerous antiviral Food and Drug Administration-approved drugs are TLR agonists. This review highlights the therapeutic approaches associated with SARS-CoV-2 and the TLRs involved in COVID-19 infection.
Collapse
Affiliation(s)
- Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | | | - Muhammad Haseeb
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
| |
Collapse
|
11
|
Ching WY, Adhikari P, Jawad B, Podgornik R. Effect of Delta and Omicron Mutations on the RBD-SD1 Domain of the Spike Protein in SARS-CoV-2 and the Omicron Mutations on RBD-ACE2 Interface Complex. Int J Mol Sci 2022; 23:10091. [PMID: 36077490 PMCID: PMC9456519 DOI: 10.3390/ijms231710091] [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: 07/29/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
The receptor-binding domain (RBD) is the essential part in the Spike-protein (S-protein) of SARS-CoV-2 virus that directly binds to the human ACE2 receptor, making it a key target for many vaccines and therapies. Therefore, any mutations at this domain could affect the efficacy of these treatments as well as the viral-cell entry mechanism. We introduce ab initio DFT-based computational study that mainly focuses on two parts: (1) Mutations effects of both Delta and Omicron variants in the RBD-SD1 domain. (2) Impact of Omicron RBD mutations on the structure and properties of the RBD-ACE2 interface system. The in-depth analysis is based on the novel concept of amino acid-amino acid bond pair units (AABPU) that reveal the differences between the Delta and/or Omicron mutations and its corresponding wild-type strain in terms of the role played by non-local amino acid interactions, their 3D shapes and sizes, as well as contribution to hydrogen bonding and partial charge distributions. Our results also show that the interaction of Omicron RBD with ACE2 significantly increased its bonding between amino acids at the interface providing information on the implications of penetration of S-protein into ACE2, and thus offering a possible explanation for its high infectivity. Our findings enable us to present, in more conspicuous atomic level detail, the effect of specific mutations that may help in predicting and/or mitigating the next variant of concern.
Collapse
Affiliation(s)
- Wai-Yim Ching
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Puja Adhikari
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Bahaa Jawad
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq
| | - Rudolf Podgornik
- School of Physical Sciences and Kavli Institute of Theoretical Science, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100090, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| |
Collapse
|
12
|
Mahmud N, Anik MI, Hossain MK, Khan MI, Uddin S, Ashrafuzzaman M, Rahaman MM. Advances in Nanomaterial-Based Platforms to Combat COVID-19: Diagnostics, Preventions, Therapeutics, and Vaccine Developments. ACS APPLIED BIO MATERIALS 2022; 5:2431-2460. [PMID: 35583460 PMCID: PMC9128020 DOI: 10.1021/acsabm.2c00123] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/24/2022] [Indexed: 12/12/2022]
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2, a ribonucleic acid (RNA) virus that emerged less than two years ago but has caused nearly 6.1 million deaths to date. Recently developed variants of the SARS-CoV-2 virus have been shown to be more potent and expanded at a faster rate. Until now, there is no specific and effective treatment for SARS-CoV-2 in terms of reliable and sustainable recovery. Precaution, prevention, and vaccinations are the only ways to keep the pandemic situation under control. Medical and scientific professionals are now focusing on the repurposing of previous technology and trying to develop more fruitful methodologies to detect the presence of viruses, treat the patients, precautionary items, and vaccine developments. Nanomedicine or nanobased platforms can play a crucial role in these fronts. Researchers are working on many effective approaches by nanosized particles to combat SARS-CoV-2. The role of a nanobased platform to combat SARS-CoV-2 is extremely diverse (i.e., mark to personal protective suit, rapid diagnostic tool to targeted treatment, and vaccine developments). Although there are many theoretical possibilities of a nanobased platform to combat SARS-CoV-2, until now there is an inadequate number of research targeting SARS-CoV-2 to explore such scenarios. This unique mini-review aims to compile and elaborate on the recent advances of nanobased approaches from prevention, diagnostics, treatment to vaccine developments against SARS-CoV-2, and associated challenges.
Collapse
Affiliation(s)
- Niaz Mahmud
- Department of Biomedical Engineering,
Military Institute of Science and Technology, Dhaka 1216,
Bangladesh
| | - Muzahidul I. Anik
- Department of Chemical Engineering,
University of Rhode Island, Kingston, Rhode Island 02881,
United States
| | - M. Khalid Hossain
- Interdisciplinary Graduate School of Engineering
Science, Kyushu University, Fukuoka 816-8580,
Japan
- Atomic Energy Research Establishment,
Bangladesh Atomic Energy Commission, Dhaka 1349,
Bangladesh
| | - Md Ishak Khan
- Department of Neurosurgery, University of
Pennsylvania, Philadelphia, Pennsylvania 19104, United
States
| | - Shihab Uddin
- Department of Applied Chemistry, Graduate School of
Engineering, Kyushu University, Fukuoka 819-0395,
Japan
- Department of Chemical Engineering,
Massachusetts Institute of Technology, Cambridge
Massachusetts 02139, United States
| | - Md. Ashrafuzzaman
- Department of Biomedical Engineering,
Military Institute of Science and Technology, Dhaka 1216,
Bangladesh
| | - Md Mushfiqur Rahaman
- Department of Emergency Medicine, NYU
Langone Health, New York, New York 10016, United
States
| |
Collapse
|
13
|
Richard D, Muthuirulan P, Aguiar J, Doxey AC, Banerjee A, Mossman K, Hirota J, Capellini TD. Intronic regulation of SARS-CoV-2 receptor (ACE2) expression mediated by immune signaling and oxidative stress pathways. iScience 2022; 25:104614. [PMID: 35756893 PMCID: PMC9213013 DOI: 10.1016/j.isci.2022.104614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 03/19/2022] [Accepted: 06/10/2022] [Indexed: 11/26/2022] Open
Abstract
The angiotensin-converting enzyme 2 (ACE2) protein is a key catalytic regulator of the renin-angiotensin system (RAS), involved in fluid homeostasis and blood pressure modulation. ACE2 also serves as a cell-surface receptor for some coronaviruses such as SARS-CoV and SARS-CoV-2. Improved characterization of ACE2 regulation may help us understand the effects of pre-existing conditions on COVID-19 incidence, as well as pathogenic dysregulation following viral infection. Here, we perform bioinformatic analyses to hypothesize on ACE2 gene regulation in two different physiological contexts, identifying putative regulatory elements of ACE2 expression. We perform functional validation of our computational predictions via targeted CRISPR-Cas9 deletions of these elements in vitro, finding them responsive to immune signaling and oxidative-stress pathways. This contributes to our understanding of ACE2 gene regulation at baseline and immune challenge. Our work supports pursuit of these putative mechanisms in our understanding of infection/disease caused by current, and future, SARS-related viruses such as SARS-CoV-2. Lung expression patterns suggest ACE2 regulation by immune and oxidative signaling CRISPR deletion of intronic regulatory elements (REs) alters ACE2 expression Effects of RE deletion are modified by immune stimulation and oxidative stress Propose two mechanisms for regulating ACE2 at baseline and after immune challenge
Collapse
Affiliation(s)
- Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138 USA
| | | | - Jennifer Aguiar
- Department of Biology, University of Waterloo, Waterloo, ON, N2L3G1 Canada
| | - Andrew C Doxey
- Department of Biology, University of Waterloo, Waterloo, ON, N2L3G1 Canada
| | - Arinjay Banerjee
- Department of Biology, University of Waterloo, Waterloo, ON, N2L3G1 Canada.,Vaccine and Infectious Disease Organization, University of Saskatchewan; Saskatoon, SK, S7N 5E3 Canada.,Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan; Saskatoon, SK, S7N5B4 Canada
| | - Karen Mossman
- Department of Medicine, McMaster University, Hamilton, ON, L8N 3Z5 Canada
| | - Jeremy Hirota
- Department of Biology, University of Waterloo, Waterloo, ON, N2L3G1 Canada.,Department of Medicine, McMaster University, Hamilton, ON, L8N 3Z5 Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, V5Z 1M9 Canada
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138 USA.,Broad Institute of MIT and Harvard, Cambridge, 02142 MA, USA
| |
Collapse
|
14
|
Karade D, Karade V. AIDrugApp: artificial intelligence-based Web-App for virtual screening of inhibitors against SARS-COV-2. J EXP THEOR ARTIF IN 2022. [DOI: 10.1080/0952813x.2022.2058619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Divya Karade
- Chemical Engineering and Process Development (Cepd) Division, CSIR-National Chemical Laboratory, Pune India
| | - Vikas Karade
- Department of Research and Development, Algosurg Products Pvt. Ltd, Mumbai, India
| |
Collapse
|
15
|
Sasidharan S, Sarkar N, Saudagar P. Discovery of compounds inhibiting SARS-COV-2 multi-targets. J Biomol Struct Dyn 2022; 41:2602-2617. [PMID: 34994297 DOI: 10.1080/07391102.2021.2025149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a pandemic that has devastated the lives of millions. Researchers around the world are relentlessly working in hopes of finding a cure. Even though the virus shares similarities with reported SARS-CoV and MERS-CoV at the genomic and proteomic level, efforts to repurpose already known drugs against SARS-CoV-2 have resulted ineffective. In this succinct review, we discuss the different potential targets in SARS-CoV-2 at both the genomic and proteomic levels. In addition, we analyze the compounds inhibiting individual target protein as well as multiple targets of SARS-CoV-2. ACE-2 receptor in humans has also been considered a target, keeping the role of the receptor in mind. The mechanism of action of these compounds has also been highlighted along with their clinical manifestation. Towards the end of the review, a brief note on the drugs currently in clinical trials and the current status of the vaccines are also examined. In conclusion, compounds targeting multiple targets of the virus hold the key in putting an end to the coronavirus malady.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| | - Neellohit Sarkar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| |
Collapse
|
16
|
Abstract
INTRODUCTION Appearances of SARS-CoV-2 variants have created havoc and additional challenges for the ongoing vaccination drive against pandemic COVID-19. Interestingly, several vaccine platforms are showing great potential to produce successful vaccines against SARS-CoV-2 and its variants. Billions of COVID-19 vaccine doses have been administered worldwide. Mix-and-Match COVID-19 vaccines involving the mixing of the same platform vaccines and also two different vaccine platforms may provide greater protection against SARS-CoV-2 and its variants. COVID-19 vaccines have become one of the most important tools to mitigate the ongoing pandemic COVID-19. AREAS COVERED We describe SARS-Cov-2 variants, their impact on the population, COVID-19 vaccines, diverse vaccine platforms, doses of vaccines, the efficacy of vaccines against SARS-CoV-2 and its variants, mitigation of the COVID-19 transmission- alternatives to vaccines. EXPERT OPINION Diverse vaccine platforms may safeguard against ongoing, deadly SARS-CoV-2 and its infectious variants. The efficacies of COVID-19 vaccines are significantly high after the administration of the second dose. Further, it protects individuals including vulnerable patients with co-morbidities from SARS-CoV-2 and its variants. The hospitalizations and deaths of the individuals may be prevented by COVID-19 vaccines.
Collapse
Affiliation(s)
- Bhaswati Chatterjee
- Chemical Science, National Institute of Pharmaceutical Education and Research, Hyderabad, India,CONTACT Bhaswati Chatterjee Chemical Science National Institute of Pharmaceutical Education and Research, India
| | - Suman S. Thakur
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, India,Suman S. Thakur Principal Scientist, Proteomics and Cell Signaling, Lab W110 Centre for Cellular and Molecular Biology Uppal Road, Hyderabad-500007, India
| |
Collapse
|
17
|
Peirouvi T, Aliaghaei A, Eslami Farsani B, Ziaeipour S, Ebrahimi V, Forozesh M, Ghadipasha M, Mahmoudiasl GR, Aryan A, Moghimi N, Abdi S, Raoofi A, Kargar Godaneh M, Abdollahifar MA. COVID-19 disrupts the blood-testis barrier through the induction of inflammatory cytokines and disruption of junctional proteins. Inflamm Res 2021; 70:1165-1175. [PMID: 34436630 PMCID: PMC8387554 DOI: 10.1007/s00011-021-01497-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Junctional proteins are the most important component of the blood-testis barrier and maintaining the integrity of this barrier is essential for spermatogenesis and male fertility. The present study elucidated the effect of SARS-CoV-2 infection on the blood-testis barrier (BTB) in patients who died from severe acute respiratory syndrome coronavirus 2 (COVID-19) complications. METHODS In this study, lung and testis tissue was collected from autopsies of COVID-19 positive (n = 10) and negative men (n = 10) and was taken for stereology, immunocytochemistry, and RNA extraction. RESULTS Evaluation of the lung tissue showed that the SARS-CoV-2 infection caused extensive damage to the lung tissue and also increases inflammation in testicular tissue and destruction of the testicular blood barrier. Autopsied testicular specimens of COVID-19 showed that COVID-19 infection significantly changes the spatial arrangement of testicular cells and notably decreased the number of Sertoli cells. Moreover, the immunohistochemistry results showed a significant reduction in the protein expression of occluding, claudin-11, and connexin-43 in the COVID-19 group. In addition, we also observed a remarkable enhancement in protein expression of CD68 in the testes of the COVID-19 group in comparison with the control group. Furthermore, the result showed that the expression of TNF-α, IL1β, and IL6 was significantly increased in COVID-19 cases as well as the expression of occludin, claudin-11, and connexin-43 was decreased in COVID-19 cases. CONCLUSIONS Overall, the present study demonstrated that SARS-CoV-2 could induce the up-regulation of the pro-inflammatory cytokine and down-regulation of junctional proteins of the BTB, which can disrupt BTB and ultimately impair spermatogenesis.
Collapse
Affiliation(s)
- Tahmineh Peirouvi
- Department of Histology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Arabi Ave, Daneshjuo Blvd, Erabi Ave, Velenjak, Tehran, Iran
| | | | - Sanaz Ziaeipour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Arabi Ave, Daneshjuo Blvd, Erabi Ave, Velenjak, Tehran, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Forozesh
- Forensic Medicine Legal Medicine Research Center, Iranian Legal Medicine Organization, Tehran, Iran
| | - Masoud Ghadipasha
- Forensic Medicine Legal Medicine Research Center, Iranian Legal Medicine Organization, Tehran, Iran
| | | | - Arefeh Aryan
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Moghimi
- Department of Histology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Arabi Ave, Daneshjuo Blvd, Erabi Ave, Velenjak, Tehran, Iran
| | - Shabnam Abdi
- Department of Anatomical Sciences and Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Raoofi
- Department of Anatomical Sciences, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mohammadhossein Kargar Godaneh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Arabi Ave, Daneshjuo Blvd, Erabi Ave, Velenjak, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Arabi Ave, Daneshjuo Blvd, Erabi Ave, Velenjak, Tehran, Iran.
| |
Collapse
|
18
|
Shirbhate E, Pandey J, Patel VK, Kamal M, Jawaid T, Gorain B, Kesharwani P, Rajak H. Understanding the role of ACE-2 receptor in pathogenesis of COVID-19 disease: a potential approach for therapeutic intervention. Pharmacol Rep 2021; 73:1539-1550. [PMID: 34176080 PMCID: PMC8236094 DOI: 10.1007/s43440-021-00303-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) and its homologue, ACE2, are commonly allied with hypertension, renin-angiotensin-aldosterone system pathway, and other cardiovascular system disorders. The recent pandemic of COVID-19 has attracted the attention of numerous researchers on ACE2 receptors, where the causative viral particle, SARS-CoV-2, is established to exploit these receptors for permitting their entry into the human cells. Therefore, studies on the molecular origin and pathophysiology of the cell response in correlation to the role of ACE2 receptors to these viruses are bringing novel theories. The varying level of manifestation and importance of ACE proteins, underlying irregularities and disorders, intake of specific medications, and persistence of assured genomic variants at the ACE genes are potential questions raising nowadays while observing the marked alteration in response to the SARS-CoV-2-infected patients. Therefore, the present review has focused on several raised opinions associated with the role of the ACE2 receptor and its impact on COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Jaiprakash Pandey
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Vijay K Patel
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box No. 173, Al-Kharj, 11942, Kingdom of Saudi Arabia
| | - Talha Jawaid
- Department of Pharmacology, College of Medicine, Al Imam Bin Saud Islamic University, Riyadh, 13314, Kingdom of Saudi Arabia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Harish Rajak
- Institute of Pharmaceutical Sciences, Guru Ghasidas University (A Central University), Bilaspur, Chhattisgarh, 495 009, India.
| |
Collapse
|
19
|
Metabolomics Signatures of SARS-CoV-2 Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:45-59. [PMID: 34735713 DOI: 10.1007/5584_2021_674] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
For a very long time, viral infections have been considered as one of the most important causes of death and disability around the world. Through the viral infection, viruses as small pathogens enter the host cells and use hosts' biosynthesis machinery to replicate and collect infectious lineages. Moreover, they can modify hosts' metabolic pathways in order to their own purposes. Nowadays (in 2019-2020), the most famous type of viral infection which was caused by a novel type of coronavirus is called COVID-19 disease. It has claimed the lives of many people around the world and is a very serious threat to health. Since investigations of the effects of viruses on host metabolism using metabolomics tools may have given focuses on novel appropriate treatments, in the current review the authors highlighted the virus-host metabolic interactions and metabolomics perspective in COVID-19.
Collapse
|
20
|
Chauhan N, Soni S, Jain U. Optimizing testing regimes for the detection of COVID-19 in children and older adults. Expert Rev Mol Diagn 2021; 21:999-1016. [PMID: 34324823 PMCID: PMC8425447 DOI: 10.1080/14737159.2021.1962708] [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: 02/22/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is a major pandemic and continuously emerging due to unclear prognosis and unavailability of reliable detection tools. Older adults are more susceptible to COVID-19 than children showing mature Angiotensin-Converting Enzyme 2 (ACE2), low concentration of immune targets, and comorbid conditions. Several detection platforms have been commercialized to date and more are in pipeline, however, the rate of false-positive results and rapid mutation of SARS-CoV-2 is increasing. Additionally, physiological, and geographical variations of affected individuals are also calling for diagnostic methods optimization.Areas Covered: Extensive information related to the optimization and usefulness of SARS-CoV-2 diagnostic methods based on sensitivity and specificity as definitive and feasible investigative tools is discussed. Moreover, an option of combining laboratory diagnostic methods to improve diagnostic strategies is also proposed and discussed in the comparative section of optimization studies.Expert Opinion: The review article explains the importance of optimization strategies for SARS-CoV-2 detection in children and older adults. There are advancements in COVID-19 detection including CRISPR-based, electrochemical, and optical-based sensing systems. However, the lack of sufficient studies on a comparative evaluation of standardized SARS-CoV-2 diagnostic methods among children and older adults, limit the authentication of commercialized kits.
Collapse
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Shringika Soni
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| |
Collapse
|
21
|
Cooper SL, Boyle E, Jefferson SR, Heslop CRA, Mohan P, Mohanraj GGJ, Sidow HA, Tan RCP, Hill SJ, Woolard J. Role of the Renin-Angiotensin-Aldosterone and Kinin-Kallikrein Systems in the Cardiovascular Complications of COVID-19 and Long COVID. Int J Mol Sci 2021; 22:8255. [PMID: 34361021 PMCID: PMC8347967 DOI: 10.3390/ijms22158255] [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] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the virus responsible for the COVID-19 pandemic. Patients may present as asymptomatic or demonstrate mild to severe and life-threatening symptoms. Although COVID-19 has a respiratory focus, there are major cardiovascular complications (CVCs) associated with infection. The reported CVCs include myocarditis, heart failure, arrhythmias, thromboembolism and blood pressure abnormalities. These occur, in part, because of dysregulation of the Renin-Angiotensin-Aldosterone System (RAAS) and Kinin-Kallikrein System (KKS). A major route by which SARS-CoV-2 gains cellular entry is via the docking of the viral spike (S) protein to the membrane-bound angiotensin converting enzyme 2 (ACE2). The roles of ACE2 within the cardiovascular and immune systems are vital to ensure homeostasis. The key routes for the development of CVCs and the recently described long COVID have been hypothesised as the direct consequences of the viral S protein/ACE2 axis, downregulation of ACE2 and the resulting damage inflicted by the immune response. Here, we review the impact of COVID-19 on the cardiovascular system, the mechanisms by which dysregulation of the RAAS and KKS can occur following virus infection and the future implications for pharmacological therapies.
Collapse
Affiliation(s)
- Samantha L. Cooper
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Eleanor Boyle
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Sophie R. Jefferson
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Calum R. A. Heslop
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Pirathini Mohan
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Gearry G. J. Mohanraj
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Hamza A. Sidow
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Rory C. P. Tan
- School of Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (E.B.); (S.R.J.); (C.R.A.H.); (P.M.); (G.G.J.M.); (H.A.S.); (R.C.P.T.)
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
- Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
22
|
Barh D, Aljabali AA, Tambuwala MM, Tiwari S, Serrano-Aroca Á, Alzahrani KJ, Silva Andrade B, Azevedo V, Ganguly NK, Lundstrom K. Predicting COVID-19-Comorbidity Pathway Crosstalk-Based Targets and Drugs: Towards Personalized COVID-19 Management. Biomedicines 2021; 9:556. [PMID: 34067609 PMCID: PMC8156524 DOI: 10.3390/biomedicines9050556] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/12/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
It is well established that pre-existing comorbid conditions such as hypertension, diabetes, obesity, cardiovascular diseases (CVDs), chronic kidney diseases (CKDs), cancers, and chronic obstructive pulmonary disease (COPD) are associated with increased severity and fatality of COVID-19. The increased death from COVID-19 is due to the unavailability of a gold standard therapeutic and, more importantly, the lack of understanding of how the comorbid conditions and COVID-19 interact at the molecular level, so that personalized management strategies can be adopted. Here, using multi-omics data sets and bioinformatics strategy, we identified the pathway crosstalk between COVID-19 and diabetes, hypertension, CVDs, CKDs, and cancers. Further, shared pathways and hub gene-based targets for COVID-19 and its associated specific and combination of comorbid conditions are also predicted towards developing personalized management strategies. The approved drugs for most of these identified targets are also provided towards drug repurposing. Literature supports the involvement of our identified shared pathways in pathogenesis of COVID-19 and development of the specific comorbid condition of interest. Similarly, shared pathways- and hub gene-based targets are also found to have potential implementations in managing COVID-19 patients. However, the identified targets and drugs need further careful evaluation for their repurposing towards personalized treatment of COVID-19 cases having pre-existing specific comorbid conditions we have considered in this analysis. The method applied here may also be helpful in identifying common pathway components and targets in other disease-disease interactions too.
Collapse
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Alaa A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK;
| | - Sandeep Tiwari
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia;
| | - Bruno Silva Andrade
- Laboratório de Bioinformática e Química Computacional, Departamento de Ciências Biológicas, Universidade Estadual do Sudoeste da Bahia (UESB), Jequié 45206-190, Brazil;
| | - Vasco Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.T.); (V.A.)
| | - Nirmal Kumar Ganguly
- National Institute of Immunology, Aruna Asaf Ali Marg, Jawaharlal Nehru University, New Delhi 110067, India;
- Institute of Liver and Biliary Science, New Delhi 110070, India
- Policy Center for Biomedical Research, Translational Health Science & Technology Institute, Faridabad 121001, India
| | | |
Collapse
|
23
|
Khan N, Kumar N, Geiger JD. Possible therapeutic targets for SARS-CoV-2 infection and COVID-19. JOURNAL OF ALLERGY AND INFECTIOUS DISEASES 2021; 2:75-83. [PMID: 37564275 PMCID: PMC10414779 DOI: 10.46439/allergy.2.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
SARS-CoV-2 infection causes COVID-19, which has emerged as a health emergency worldwide. SARS-CoV-2 infects cells by binding to ACE2 receptors and enters into the cytoplasm following its escape from endolysosomes. Once in the cytoplasm, the virus replicates and eventually causes various pathological conditions including acute respiratory distress syndrome (ARDS) that is caused by pro-inflammatory cytokine storms. Thus, endolysosomes and cytokine storms are important therapeutic targets to suppress SARS-CoV-2 infection and COVID-19. Here, we discuss therapeutic targets of SARS-CoV-2 infection and available drugs that could be helpful in the suppression of the SARS-CoV-2 infection and pathological condition COVID-19. The urgency of the COVID-19 pandemic precludes the development of new drugs and increased focus on drug repurposing might provide the quickest way to finding effective medicines.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Nirmal Kumar
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|