1
|
Robert Kolar M, Mitra D, Kobzarenko V. Efficient discovery of frequently co-occurring mutations in a sequence database with matrix factorization. PLoS Comput Biol 2025; 21:e1012391. [PMID: 40273414 DOI: 10.1371/journal.pcbi.1012391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
We have developed a robust method for efficiently tracking multiple co-occurring mutations in a sequence database. Evolution often hinges on the interaction of several mutations to produce significant phenotypic changes that lead to the proliferation of a variant. However, identifying numerous simultaneous mutations across a vast database of sequences poses a significant computational challenge. Our approach leverages a matrix factorization technique to automatically and efficiently pinpoint subsets of positions where co-mutations occur, appearing in a substantial number of sequences within the database. We validated our method using SARS-CoV-2 receptor-binding domains, comprising approximately seven hundred thousand sequences of the Spike protein, demonstrating superior performance compared to a reasonably exhaustive brute-force method. Furthermore, we explore the biological significance of the identified co-mutational positions (CMPs) and their potential impact on the virus's evolution and functionality, identifying key mutations in Delta and Omicron variants. This analysis underscores the significant role of identified CMPs in understanding the evolutionary trajectory. By tracking the "birth" and "death" of CMPs, we can elucidate the persistence and impact of specific groups of mutations across different viral strains, providing valuable insights into the virus' adaptability and thus, possibly aiding vaccine design strategies.
Collapse
Affiliation(s)
- Michael Robert Kolar
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| | - Debasis Mitra
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| | - Valerie Kobzarenko
- BiC Lab, Department of Electrical Engineering and Computer Science, Florida Institute of Technology, Melbourne, Florida, United States of America
| |
Collapse
|
2
|
Saita T, Thitanuwat B, Niyomdecha N, Prasertsopon J, Lerdsamran H, Puthavathana P, Noisumdaeng P. Measuring SARS-CoV-2 RNA in Bangkok wastewater treatment plants and estimating infected population after fully opening the country in 2023, Thailand. Sci Rep 2025; 15:9663. [PMID: 40113890 PMCID: PMC11926235 DOI: 10.1038/s41598-025-94938-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/18/2025] [Indexed: 03/22/2025] Open
Abstract
Wastewater-based epidemiology (WBE) has been employed for monitoring the presence of SARS-CoV-2 infected population. Herein, the study aims to apply the WBE for surveillance and monitoring SARS-CoV-2 in Bangkok, where the highest official covid-19 cases reported in Thailand, during the fully opening for international tourists in early 2023. A total of 200 wastewater samples (100 influent and 100 effluent samples) were collected from 10 wastewater treatment plants (WWTPs) during January-May 2023. SARS-CoV-2 RNA was detected by real time qRT-PCR with accounting for 51% (102/200). Of these, 88% (88/100) and 14% (14/100) were detected in influent and effluent samples, respectively. The SARS-CoV-2 RNA concentration was detected in ranged of 4.76 × 102-1.48 × 105 copies/L. The amount of SARS-CoV-2 RNA has increased approximately 4 times from the lag phase (January-March) to the log phase (April-May). Spearman's correlation coefficient revealed that correlation between estimated infected population and weekly reported cases was statistically significant (p-value = 0.017). SARS-CoV-2 RNA in influent had a statistically significant relationship with weekly reported cases (r = 0.481, p-value < 0.001). Lag time analysis revealed early warning 1-3 weeks before rising covid-19 cases observed. GIS was applied for spatial-temporal analysis at the province level, suggesting real time dashboard should be further developed.
Collapse
Affiliation(s)
- Thanchira Saita
- Faculty of Public Health, Thammasat University, Pathum Thani, 12121, Thailand
- Thammasat University Research Unit in Modern Microbiology and Public Health Genomics, Thammasat University, Pathum Thani, 12121, Thailand
| | | | - Nattamon Niyomdecha
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12121, Thailand
| | - Jarunee Prasertsopon
- Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Hatairat Lerdsamran
- Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | | | - Pirom Noisumdaeng
- Faculty of Public Health, Thammasat University, Pathum Thani, 12121, Thailand.
- Thammasat University Research Unit in Modern Microbiology and Public Health Genomics, Thammasat University, Pathum Thani, 12121, Thailand.
| |
Collapse
|
3
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
4
|
Blanco-Agudín N, Ye S, González-Fernández S, Alcalde I, Merayo-Lloves J, Quirós LM. Exosomes in Ocular Health: Recent Insights into Pathology, Diagnostic Applications and Therapeutic Functions. Biomedicines 2025; 13:233. [PMID: 39857816 PMCID: PMC11762739 DOI: 10.3390/biomedicines13010233] [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: 11/28/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Exosomes are extracellular vesicles ranging from 30 to 150 nm in diameter that contain proteins, nucleic acids and other molecules. Produced by virtually all cell types, they travel throughout the body until they reach their target, where they can trigger a wide variety of effects by transferring the molecular cargo to recipient cells. In the context of ocular physiology, exosomes play a very important role in embryological development, the regulation of homeostasis and the immune system, which is crucial for normal vision. Consequently, in pathological situations, exosomes also undergo modifications in terms of quantity, composition and content, depending on the etiology of the disease. However, the mechanisms by which exosomes contribute to ocular pathology has not yet been studied in depth, and many questions remain unanswered. This review aims to summarize the most recent knowledge on the function of exosomes in the ocular system in healthy individuals and the role they play during pathological processes of a degenerative, infectious, neurodegenerative, vascular and inflammatory nature, such as keratoconus, keratitis, glaucoma, diabetic retinopathy and uveitis. Furthermore, given their unique characteristics, their potential as diagnostic biomarkers or therapeutic agents and their application in clinical ophthalmology are also explored, along with the main limitations that researchers face today in the field.
Collapse
Affiliation(s)
- Noelia Blanco-Agudín
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Suhui Ye
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Sara González-Fernández
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Luis M. Quirós
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
5
|
Balzanelli MG, Rastmanesh R, Distratis P, Lazzaro R, Inchingolo F, Del Prete R, Pham VH, Aityan SK, Cong TT, Nguyen KCD, Isacco CG. The Role of SARS-CoV-2 Spike Protein in Long-term Damage of Tissues and Organs, the Underestimated Role of Retrotransposons and Stem Cells, a Working Hypothesis. Endocr Metab Immune Disord Drug Targets 2025; 25:85-98. [PMID: 38468535 DOI: 10.2174/0118715303283480240227113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 03/13/2024]
Abstract
Coronavirus disease-2019 (COVID-19) is a respiratory disease in which Spike protein from SARS-CoV-2 plays a key role in transferring virus genomic code into target cells. Spike protein, which is found on the surface of the SARS-CoV-2 virus, latches onto angiotensin-converting enzyme 2 receptors (ACE2r) on target cells. The RNA genome of coronaviruses, with an average length of 29 kb, is the longest among all RNA viruses and comprises six to ten open reading frames (ORFs) responsible for encoding replicase and structural proteins for the virus. Each component of the viral genome is inserted into a helical nucleocapsid surrounded by a lipid bilayer. The Spike protein is responsible for damage to several organs and tissues, even leading to severe impairments and long-term disabilities. Spike protein could also be the cause of the long-term post-infectious conditions known as Long COVID-19, characterized by a group of unresponsive idiopathic severe neuro- and cardiovascular disorders, including strokes, cardiopathies, neuralgias, fibromyalgia, and Guillaume- Barret's like-disease. In this paper, we suggest a pervasive mechanism whereby the Spike proteins either from SARS-CoV-2 mRNA or mRNA vaccines, tend to enter the mature cells, and progenitor, multipotent, and pluripotent stem cells (SCs), altering the genome integrity. This will eventually lead to the production of newly affected clones and mature cells. The hypothesis presented in this paper proposes that the mRNA integration into DNA occurs through several components of the evolutionarily genetic mechanism such as retrotransposons and retrotransposition, LINE-1 or L1 (long interspersed element-1), and ORF-1 and 2 responsible for the generation of retrogenes. Once the integration phase is concluded, somatic cells, progenitor cells, and SCs employ different silencing mechanisms. DNA methylation, followed by histone modification, begins to generate unlimited lines of affected cells and clones that form affected tissues characterized by abnormal patterns that become targets of systemic immune cells, generating uncontrolled inflammatory conditions, as observed in both Long COVID-19 syndrome and the mRNA vaccine.
Collapse
Affiliation(s)
- Mario G Balzanelli
- 118 SET, Department of Pre-hospital and Emergency, SG Giuseppe Moscati Hospital, 74120 Taranto, Italy
| | - Reza Rastmanesh
- Department of Nutrition and Metabolism, The Nutrition Society, Boyd Orr House, 10 Cambridge Court, 210 Shepherds Bush Road, London, UK
| | - Pietro Distratis
- 118 SET, Department of Pre-hospital and Emergency, SG Giuseppe Moscati Hospital, 74120 Taranto, Ital
| | - Rita Lazzaro
- 118 SET, Department of Pre-hospital and Emergency, SG Giuseppe Moscati Hospital, 74120 Taranto, Ital
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Raffaele Del Prete
- Department of Interdisciplinary Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Van H Pham
- Phan Chau Trinh University of Medicine, Quang Nam 70000, Vietnam
| | - Sergey K Aityan
- Northwestern University, Multidisciplinary Research Center, Oakland, CA 94612, USA
| | - Toai Tran Cong
- Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| | - Kieu C D Nguyen
- Department of Interdisciplinary Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Ciro Gargiulo Isacco
- 118 SET, Department of Pre-hospital and Emergency, SG Giuseppe Moscati Hospital, 74120 Taranto, Italy
- Department of Interdisciplinary Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
6
|
Hsiung KC, Chiang HJ, Reinig S, Shih SR. Vaccine Strategies Against RNA Viruses: Current Advances and Future Directions. Vaccines (Basel) 2024; 12:1345. [PMID: 39772007 PMCID: PMC11679499 DOI: 10.3390/vaccines12121345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The development of vaccines against RNA viruses has undergone a rapid evolution in recent years, particularly driven by the COVID-19 pandemic. This review examines the key roles that RNA viruses, with their high mutation rates and zoonotic potential, play in fostering vaccine innovation. We also discuss both traditional and modern vaccine platforms and the impact of new technologies, such as artificial intelligence, on optimizing immunization strategies. This review evaluates various vaccine platforms, ranging from traditional approaches (inactivated and live-attenuated vaccines) to modern technologies (subunit vaccines, viral and bacterial vectors, nucleic acid vaccines such as mRNA and DNA, and phage-like particle vaccines). To illustrate these platforms' practical applications, we present case studies of vaccines developed for RNA viruses such as SARS-CoV-2, influenza, Zika, and dengue. Additionally, we assess the role of artificial intelligence in predicting viral mutations and enhancing vaccine design. The case studies underscore the successful application of RNA-based vaccines, particularly in the fight against COVID-19, which has saved millions of lives. Current clinical trials for influenza, Zika, and dengue vaccines continue to show promise, highlighting the growing efficacy and adaptability of these platforms. Furthermore, artificial intelligence is driving improvements in vaccine candidate optimization and providing predictive models for viral evolution, enhancing our ability to respond to future outbreaks. Advances in vaccine technology, such as the success of mRNA vaccines against SARS-CoV-2, highlight the potential of nucleic acid platforms in combating RNA viruses. Ongoing trials for influenza, Zika, and dengue demonstrate platform adaptability, while artificial intelligence enhances vaccine design by predicting viral mutations. Integrating these innovations with the One Health approach, which unites human, animal, and environmental health, is essential for strengthening global preparedness against future RNA virus threats.
Collapse
Affiliation(s)
- Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
7
|
Alqahtani SAM. Mucosal immunity in COVID-19: a comprehensive review. Front Immunol 2024; 15:1433452. [PMID: 39206184 PMCID: PMC11349522 DOI: 10.3389/fimmu.2024.1433452] [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: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Mucosal immunity plays a crucial role in defending against coronaviruses, particularly at respiratory sites, serving as the first line of defense against viral invasion and replication. Coronaviruses have developed various immune evasion strategies at the mucosal immune system, hindering the recognition of infected cells and evading antibody responses. Understanding the immune mechanisms and responses is crucial for developing effective vaccines and therapeutics against coronaviruses. The role of mucosal immunity in COVID-19 is significant, influencing both local and systemic immune responses to the virus. Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Mucosally delivered vaccines and those under clinical trials are being compared and contrasted to understand their effectiveness in inducing mucosal immunity against coronaviruses. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community-acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality. This comprehensive review article outlines the current evidence about the role of mucosal immune responses in the clearance of SARS-CoV-2 infection, as well as potential mucosal mechanisms of protection against (re-)infection. It also proposes that there is a significant role for mucosal immunity and for secretory as well as circulating IgA antibodies in COVID-19, and that it is important to elucidate this in order to comprehend especially the asymptomatic and mild states of the infection, which appear to account for the majority of cases. Moreover, it is possible that mucosal immunity can be exploited for beneficial diagnostic, therapeutic, or prophylactic purposes. The findings from recent studies on mucosal immunity in COVID-19 can be used to develop effective vaccines and treatments that can effectively target both mucosal and systemic immune responses.
Collapse
|
8
|
Valiate BVS, Castro JTD, Marçal TG, Andrade LAF, Oliveira LID, Maia GBF, Faustino LP, Hojo-Souza NS, Reis MAAD, Bagno FF, Salazar N, Teixeira SR, Almeida GG, Gazzinelli RT. Evaluation of an RBD-nucleocapsid fusion protein as a booster candidate for COVID-19 vaccine. iScience 2024; 27:110177. [PMID: 38993669 PMCID: PMC11238127 DOI: 10.1016/j.isci.2024.110177] [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: 08/17/2023] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Despite successful vaccines and updates, constant mutations of SARS-CoV-2 makes necessary the search for new vaccines. We generated a chimeric protein that comprises the receptor-binding domain from spike and the nucleocapsid antigens (SpiN) from SARS-CoV-2. Once SpiN elicits a protective immune response in rodents, here we show that convalescent and previously vaccinated individuals respond to SpiN. CD4+ and CD8+ T cells from these individuals produced greater amounts of IFN-γ when stimulated with SpiN, compared to SARS-CoV-2 antigens. Also, B cells from these individuals were able to secrete antibodies that recognize SpiN. When administered as a boost dose in mice previously immunized with CoronaVac, ChAdOx1-S or BNT162b2, SpiN was able to induce a greater or equivalent immune response to homologous prime/boost. Our data reveal the ability of SpiN to induce cellular and humoral responses in vaccinated human donors, rendering it a promising candidate.
Collapse
Affiliation(s)
- Bruno Vinicius Santos Valiate
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Julia Teixeira de Castro
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Luis Adan Flores Andrade
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Livia Isabela de Oliveira
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte 31.630-901, MG, Brazil
| | | | | | - Natalia S Hojo-Souza
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Natalia Salazar
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Gregório Guilherme Almeida
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Ricardo Tostes Gazzinelli
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| |
Collapse
|
9
|
Kang L, Wahaab A, Qi P, Qiu Y, Wei J, Li B, Shao D, Li Z, Liu K, Ma Z, Su S. Porcine reproductive and respiratory syndrome virus nsp4-mediated β2M downregulation contributes to SLA-I decrease and virus infection in vivo and in vitro. Virology 2024; 595:110083. [PMID: 38696887 DOI: 10.1016/j.virol.2024.110083] [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: 11/30/2023] [Revised: 03/09/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) infection inhibits swine leukocyte antigen class I (SLA-I) expression in pigs, resulting in inefficient antigen presentation and subsequent low levels of cellular PRRSV-specific immunity as well as persistent viremia. We previously observed that the non-structural protein 4 (nsp4) of PRRSV contributed to inhibition of the β2-microglobulin (β2M) and SLA-I expression in cells. Here, we constructed a series of nsp4 mutants with different combination of amino acid mutations to attenuate the inhibitory effect of nsp4 on β2M and SLA-I expression. Almost all nsp4 mutants exogenously expressed in cells showed an attenuated effect on inhibition of β2M and SLA-I expression, but the recombinant PRRSV harboring these nsp4 mutants failed to be rescued with exception of the rPRRSV-nsp4-mut10 harboring three amino acid mutations. However, infection of rPRRSV-nsp4-mut10 not only enhanced β2M and SLA-I expression in both cells and pigs but also promoted the DCs to active the CD3+CD8+T lymphocytes more efficiently, as compared with its parental PRRSV (rPRRVS-nsp4-wt). These data suggested that the inhibition of nsp4-mediated β2M downregulation improved β2M/SLA-I expression in pigs.
Collapse
Affiliation(s)
- Lei Kang
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Abdul Wahaab
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Pengfei Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Zongjie Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China.
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Shanghai, 200241, China.
| | - Shuo Su
- MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
10
|
Liang L, Wang B, Zhang Q, Zhang S, Zhang S. Antibody drugs targeting SARS-CoV-2: Time for a rethink? Biomed Pharmacother 2024; 176:116900. [PMID: 38861858 DOI: 10.1016/j.biopha.2024.116900] [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: 02/14/2024] [Revised: 04/20/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) heavily burdens human health. Multiple neutralizing antibodies (nAbs) have been issued for emergency use or tested for treating infected patients in the clinic. However, SARS-CoV-2 variants of concern (VOC) carrying mutations reduce the effectiveness of nAbs by preventing neutralization. Uncoding the mutation profile and immune evasion mechanism of SARS-CoV-2 can improve the outcome of Ab-mediated therapies. In this review, we first outline the development status of anti-SARS-CoV-2 Ab drugs and provide an overview of SARS-CoV-2 variants and their prevalence. We next focus on the failure causes of anti-SARS-CoV-2 Ab drugs and rethink the design strategy for developing new Ab drugs against COVID-19. This review provides updated information for the development of therapeutic Ab drugs against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Likeng Liang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bo Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Qing Zhang
- Department of Laboratory Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
11
|
Schwartz J, Capistrano KJ, Gluck J, Hezarkhani A, Naqvi AR. SARS-CoV-2, periodontal pathogens, and host factors: The trinity of oral post-acute sequelae of COVID-19. Rev Med Virol 2024; 34:e2543. [PMID: 38782605 PMCID: PMC11260190 DOI: 10.1002/rmv.2543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
COVID-19 as a pan-epidemic is waning but there it is imperative to understand virus interaction with oral tissues and oral inflammatory diseases. We review periodontal disease (PD), a common inflammatory oral disease, as a driver of COVID-19 and oral post-acute-sequelae conditions (PASC). Oral PASC identifies with PD, loss of teeth, dysgeusia, xerostomia, sialolitis-sialolith, and mucositis. We contend that PD-associated oral microbial dysbiosis involving higher burden of periodontopathic bacteria provide an optimal microenvironment for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These pathogens interact with oral epithelial cells activate molecular or biochemical pathways that promote viral adherence, entry, and persistence in the oral cavity. A repertoire of diverse molecules identifies this relationship including lipids, carbohydrates and enzymes. The S protein of SARS-CoV-2 binds to the ACE2 receptor and is activated by protease activity of host furin or TRMPSS2 that cleave S protein subunits to promote viral entry. However, PD pathogens provide additional enzymatic assistance mimicking furin and augment SARS-CoV-2 adherence by inducing viral entry receptors ACE2/TRMPSS, which are poorly expressed on oral epithelial cells. We discuss the mechanisms involving periodontopathogens and host factors that facilitate SARS-CoV-2 infection and immune resistance resulting in incomplete clearance and risk for 'long-haul' oral health issues characterising PASC. Finally, we suggest potential diagnostic markers and treatment avenues to mitigate oral PASC.
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Oral Medicine and Diagnostic Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | | | - Joseph Gluck
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Armita Hezarkhani
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Afsar R. Naqvi
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| |
Collapse
|
12
|
Haque A, Pant AB. The coevolution of Covid-19 and host immunity. EXPLORATION OF MEDICINE 2024:167-184. [DOI: 10.37349/emed.2024.00214] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/15/2024] [Indexed: 01/05/2025] Open
Abstract
The dynamic of the virus-host interaction is subject to constant
evolution, which makes it difficult to predict when the SARS-CoV-2 pandemic
will become endemic. Vaccines in conjunction with efforts around masking and
social distancing have reduced SARS-CoV-2 infection rates, however, there
are still significant challenges to contend with before the pandemic shifts
to endemic, such as the coronavirus acquiring mutations that allow the virus
to dodge the immunity acquired by hosts. SARS-CoV-2 variants deploy
convergent evolutionary mechanisms to sharpen their ability to impede the
host’s innate immune response. The continued emergence of variants and
sub-variants poses a significant hurdle to reaching endemicity. This
underscores the importance of continued public health measures to control
SARS-CoV-2 transmission and the need to develop better second-generation
vaccines and effective treatments that would tackle current and future
variants. We hypothesize that the hosts’ immunity to the virus is also
evolving, which is likely to abet the process of reaching
endemicity.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Geisel School of
Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | |
Collapse
|
13
|
Vajdi M, Karimi A, Hassanizadeh S, Farhangi MA, Bagherniya M, Askari G, Roufogalis BD, Davies NM, Sahebkar A. Effect of polyphenols against complications of COVID-19: current evidence and potential efficacy. Pharmacol Rep 2024; 76:307-327. [PMID: 38498260 DOI: 10.1007/s43440-024-00585-6] [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: 09/23/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
The COVID-19 pandemic that started in 2019 and resulted in significant morbidity and mortality continues to be a significant global health challenge, characterized by inflammation, oxidative stress, and immune system dysfunction.. Developing therapies for preventing or treating COVID-19 remains an important goal for pharmacology and drug development research. Polyphenols are effective against various viral infections and can be extracted and isolated from plants without losing their therapeutic potential. Researchers have developed methods for separating and isolating polyphenols from complex matrices. Polyphenols are effective in treating common viral infections, including COVID-19, and can also boost immunity. Polyphenolic-based antiviral medications can mitigate SARS-CoV-2 enzymes vital to virus replication and infection. Individual polyphenolic triterpenoids, flavonoids, anthraquinonoids, and tannins may also inhibit the SARS-CoV-2 protease. Polyphenol pharmacophore structures identified to date can explain their action and lead to the design of novel anti-COVID-19 compounds. Polyphenol-containing mixtures offer the advantages of a well-recognized safety profile with few known severe side effects. However, studies to date are limited, and further animal studies and randomized controlled trials are needed in future studies. The purpose of this study was to review and present the latest findings on the therapeutic impact of plant-derived polyphenols on COVID-19 infection and its complications. Exploring alternative approaches to traditional therapies could aid in developing novel drugs and remedies against coronavirus infection.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shirin Hassanizadeh
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Basil D Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Montenegro AFL, Clementino MAF, Yaochite JNU. Type I interferon pathway genetic variants in severe COVID-19. Virus Res 2024; 342:199339. [PMID: 38354910 PMCID: PMC10901847 DOI: 10.1016/j.virusres.2024.199339] [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: 11/21/2023] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Coronavirus Disease 2019 (COVID-19) is an infectious disease caused by SARS-CoV-2. According to the World Health Organization (WHO), there have been over 760 million reported cases and over 6 million deaths caused by this disease worldwide. The severity of COVID-19 is based on symptoms presented by the patient and is divided as asymptomatic, mild, moderate, severe, and critical. The manifestations are interconnected with genetic variations. The innate immunity is the quickest response mechanism of an organism against viruses. Type I interferon pathway plays a key role in antiviral responses due to viral replication inhibition in infected cells and adaptive immunity stimulation induced by interferon molecules. Thus, variants in type I interferon pathway's genes are being studied in different COVID-19 manifestations. This review summarizes the role of variants in type I interferon pathway's genes on prognosis and severity progression of COVID-19.
Collapse
Affiliation(s)
- A F L Montenegro
- Laboratório de Imunologia Celular e Molecular, Departamento de Análises Clínicas e Toxicológicas da Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará - UFC, Rua Pastor Samuel Munguba, 1210 - Rodolfo Teófilo, Fortaleza, Ceará, Brasil
| | - M A F Clementino
- Laboratório de Toxinologia Molecular, NUBIMED - Núcleo de Biomedicina, Universidade Federal do Ceará - UFC. Fortaleza, Ceará, Brasil
| | - J N U Yaochite
- Laboratório de Imunologia Celular e Molecular, Departamento de Análises Clínicas e Toxicológicas da Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará - UFC, Rua Pastor Samuel Munguba, 1210 - Rodolfo Teófilo, Fortaleza, Ceará, Brasil.
| |
Collapse
|
15
|
Zhou C, Huang M, Wang S, Chu S, Zhang Z, Chen N. Tunneling nanotubes: The transport highway for astrocyte-neuron communication in the central nervous system. Brain Res Bull 2024; 209:110921. [PMID: 38447659 DOI: 10.1016/j.brainresbull.2024.110921] [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: 01/02/2024] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Tunneling nanotubes (TNTs) have emerged as pivotal structures for intercellular communication, enabling the transfer of cellular components across distant cells. Their involvement in neurological disorders has attracted considerable scientific interest. This review delineates the functions of TNTs within the central nervous system, examining their role in the transmission of bioenergetic substrates, and signaling molecules, and their multifaceted impact on both physiological and pathological processes, with an emphasis on neurodegenerative diseases. The review highlights the selectivity and specificity of TNTs as dedicated pathways for intercellular cargo delivery, particularly under stress conditions that provoke increased TNT formation. The potential of TNTs as therapeutic targets is explored in depth. We pay particular attention to the interactions between astrocytes and neurons mediated by TNTs, which are fundamental to brain architecture and function. Dysfunctions in these interactions are implicated in the spread of protein aggregates and mitochondrial anomalies, contributing to the pathogenesis of neurodegenerative diseases. The review culminates with a synthesis of the current understanding of TNT biology and identifies research gaps, advocating for intensified exploration into TNTs as a promising therapeutic frontier.
Collapse
Affiliation(s)
- Cuixiang Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Min Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shasha Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
16
|
Aljabali AAA, El-Tanani M, Barh D, Tambuwala MM. COVID-19: Perspectives on innate immune evasion. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 213:171-214. [PMID: 40246344 DOI: 10.1016/bs.pmbts.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The ongoing global health challenges posed by the SARS-CoV-2, the virus responsible for the COVID-19 pandemic, necessitate a deep understanding of its intricate strategies to evade the innate immune system. This chapter aims to provide insights into the sophisticated mechanisms employed by SARS-CoV-2 in its interaction with pattern recognition receptors (PRRs), with particular emphasis on Toll-like receptors (TLRs) and RIG-I-like receptors (RLRs). By skillfully circumventing these pivotal components, the virus manages to elude detection and impairs the initiation of crucial antiviral immune responses. A notable aspect of SARS-CoV-2's immune evasion tactics lies in its strategic manipulation of cytokine production. This orchestrated modulation disrupts the delicate balance of inflammation, potentially leading to severe complications, including the notorious cytokine storm. In this regard, key viral proteins, such as the spike protein and nucleocapsid protein, emerge as pivotal players in the immune evasion process, further highlighting their significance in the context of COVID-19 pathogenesis. Acquiring a comprehensive understanding of these intricate immune evasion mechanisms holds immense promise for the development of effective treatments against COVID-19. Moreover, it is imperative for vaccine development to consider these evasion strategies to maximize vaccine efficacy. Future therapeutic interventions may involve targeting alternative pathways or augmenting the antiviral immune responses, thereby mitigating the impact of immune evasion, and fostering successful outcomes. By unraveling the underlying mechanisms of innate immune evasion, we advance our comprehension of COVID-19 pathogenesis and pave the way for the development of innovative therapeutic strategies. This comprehensive understanding catalyzes progress, enabling researchers and clinicians to devise novel approaches that combat the challenges posed by SARS-CoV-2 and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan.
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics & Applied Biotechnology, Purba Medinipur, West Bengal, India; Department of Genetics, Ecology & Evolution, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, United Kingdom.
| |
Collapse
|
17
|
Lv W, Li Z, Wang S, He J, Zhang L. A role for tunneling nanotubes in virus spread. Front Microbiol 2024; 15:1356415. [PMID: 38435698 PMCID: PMC10904554 DOI: 10.3389/fmicb.2024.1356415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Tunneling nanotubes (TNTs) are actin-rich intercellular conduits that mediate distant cell-to-cell communication and enable the transfer of various cargos, including proteins, organelles, and virions. They play vital roles in both physiological and pathological processes. In this review, we focus on TNTs in different types of viruses, including retroviruses such as HIV, HTLV, influenza A, herpesvirus, paramyxovirus, alphavirus and SARS-CoV-2. We summarize the viral proteins responsible for inducing TNT formation and explore how these virus-induced TNTs facilitate intercellular communication, thereby promoting viral spread. Furthermore, we highlight other virus infections that can induce TNT-like structures, facilitating the dissemination of viruses. Moreover, TNTs promote intercellular spread of certain viruses even in the presence of neutralizing antibodies and antiviral drugs, posing significant challenges in combating viral infections. Understanding the mechanisms underlying viral spread via TNTs provides valuable insights into potential drug targets and contributes to the development of effective therapies for viral infections.
Collapse
Affiliation(s)
- Weimiao Lv
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zichen Li
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shule Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Jingyi He
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
18
|
D’Avila H, Lima CNR, Rampinelli PG, Mateus LCO, de Sousa Silva RV, Correa JR, de Almeida PE. Lipid Metabolism Modulation during SARS-CoV-2 Infection: A Spotlight on Extracellular Vesicles and Therapeutic Prospects. Int J Mol Sci 2024; 25:640. [PMID: 38203811 PMCID: PMC10778989 DOI: 10.3390/ijms25010640] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) have a significant impact on the pathophysiological processes associated with various diseases such as tumors, inflammation, and infection. They exhibit molecular, biochemical, and entry control characteristics similar to viral infections. Viruses, on the other hand, depend on host metabolic machineries to fulfill their biosynthetic requirements. Due to potential advantages such as biocompatibility, biodegradation, and efficient immune activation, EVs have emerged as potential therapeutic targets against the SARS-CoV-2 infection. Studies on COVID-19 patients have shown that they frequently have dysregulated lipid profiles, which are associated with an increased risk of severe repercussions. Lipid droplets (LDs) serve as organelles with significant roles in lipid metabolism and energy homeostasis as well as having a wide range of functions in infections. The down-modulation of lipids, such as sphingolipid ceramide and eicosanoids, or of the transcriptional factors involved in lipogenesis seem to inhibit the viral multiplication, suggesting their involvement in the virus replication and pathogenesis as well as highlighting their potential as targets for drug development. Hence, this review focuses on the role of modulation of lipid metabolism and EVs in the mechanism of immune system evasion during SARS-CoV-2 infection and explores the therapeutic potential of EVs as well as application for delivering therapeutic substances to mitigate viral infections.
Collapse
Affiliation(s)
- Heloisa D’Avila
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | | | - Pollianne Garbero Rampinelli
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Laiza Camila Oliveira Mateus
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - Renata Vieira de Sousa Silva
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, University of Brasília, Brasília 70910-900, Brazil;
| | - Patrícia Elaine de Almeida
- Cell Biology Laboratory, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (H.D.); (P.G.R.); (L.C.O.M.); (R.V.d.S.S.)
| |
Collapse
|
19
|
Ray SK, Mukherjee S. A New-fangled COVID-19 Variant, Eris, Might be the One to Lookout in 2023 or far from Over. Infect Disord Drug Targets 2024; 24:e220124225916. [PMID: 38258765 DOI: 10.2174/0118715265276833240105110046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/12/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024]
Abstract
Coronavirus Disease-19 (COVID-19) is an infectious disease brought on by the extremely pathogenic and contagious severe acute respiratory syndrome-virus-2 (SARS-CoV-2). The agenda for the COVID-19 pandemic is dynamic and includes recent developments. Seven variants under monitoring (VUMs), one variant of interest (VOI), XBB.1.5, and their offspring lineages are currently being actively monitored by WHO. The VUMs are BA.2.75, CH.1.1, BQ.1, XBB (with the exception of XBB.1.5, XBB.1.16, and XBB.1.9.1), XBF, and XBB.1.16. With 95 countries having reported finding XBB.1.5 (VOI), it is still the most common strain worldwide, responsible for 47.9% of cases from epidemiological January to March 2023. Seventy nations discovered XBB.1.5 in February and March 2023 and posted sequencing data to GISAID. Of the 43 nations that uploaded more than 50 sequences, XBB.1.5 prevalence has increased to more than 50% in 11 nations. Over 23000 deaths and 3 million new cases were recorded globally in March and April 2023. Worldwide detection of a new COVID-19 strain has prompted specialists to issue a warning that the virus is "circulating unchecked". The Greek goddess of conflict and discord Eris has inspired the nicknaming of EG 5.1, a subvariant of Omicron. The strain is becoming more prevalent in the USA and cases are increasing in the UK. The severity of each SARS-CoV- 2 variant has been comparable, although a more severe form might develop. Eris is an ancestor of Omicron and exhibits some of its characteristics. Reinfection risk can be influenced by a variety of variables, including age, location, and health equity and the COVID-19 vaccine is more or less effective depending on the strain.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh, 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, 462020, India
| |
Collapse
|
20
|
El-Maradny YA, Rubio-Casillas A, Mohamed KI, Uversky VN, Redwan EM. Intrinsic factors behind long-COVID: II. SARS-CoV-2, extracellular vesicles, and neurological disorders. J Cell Biochem 2023; 124:1466-1485. [PMID: 37801299 DOI: 10.1002/jcb.30486] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
With the decline in the number of new Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infections, the World Health Organization announced the end of the SARS-CoV-2 pandemic. However, the repercussions of this viral pandemic may remain with us for a longer period of time, as it has remodeled the lives of humankind in many ways, including social and economic. Of course, its most important repercussions remain on the human health level. Long-coronavirus disease (COVID) or post-COVID is a state for which we do not have a concrete definition, a specific international classification of diseases Code, clear diagnostic tools, or well-known effective cures as of yet. In this second article from the Intrinsic Factors behind long-COVID Series, we try to link long-COVID symptoms with their causes, starting from the nervous system. Extracellular vesicles (ECVs) play very complex and ramified roles in the bodies of both healthy and not-healthy individuals. ECVs may facilitate the entry of many bioactive molecules and pathogens into the tissues and cells of the nervous system across the blood-brain barrier. Based on the size, quantity, and quality of their cargo, ECVs are directly proportional to the pathological condition and its severity through intertwined mechanisms that evoke inflammatory immune responses typically accompanied by pathological symptoms over variable time periods according to the type of these symptoms.
Collapse
Affiliation(s)
- Yousra A El-Maradny
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Egypt
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein, Egypt
| | - Alberto Rubio-Casillas
- Biology Laboratory, Autlán Regional Preparatory School, University of Guadalajara, Autlán, Jalisco, Mexico
| | - Kareem I Mohamed
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
21
|
Rubio-Casillas A, Redwan EM, Uversky VN. Does SARS-CoV-2 Induce IgG4 Synthesis to Evade the Immune System? Biomolecules 2023; 13:1338. [PMID: 37759738 PMCID: PMC10526126 DOI: 10.3390/biom13091338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2, the virus that causes the COVID-19 disease, has been shown to cause immune suppression in certain individuals. This can manifest as a reduced ability of the host's immune system to effectively control the infection. Studies have reported that patients with COVID-19 can exhibit a decline in white blood cell counts, including natural killer cells and T cells, which are integral components of the immune system's response to viral pathogens. These cells play critical roles in the immune response to viral infections, and their depletion can make it harder for the body to mount an effective defense against the virus. Additionally, the virus can also directly infect immune cells, further compromising their ability to function. Some individuals with severe COVID-19 pneumonia may develop a "cytokine storm", an overactive immune response that may result in tissue damage and organ malfunction. The underlying mechanisms of immune suppression in SARS-CoV-2 are not entirely understood at this time, and research is being conducted to gain a more comprehensive understanding. Research has shown that severe SARS-CoV-2 infection promotes the synthesis of IgG4 antibodies. In this study, we propose the hypothesis that IgG4 antibodies produced by B cells in response to infection by SARS-CoV-2 generate immunological tolerance, which prevents its elimination and leads to persistent and chronic infection. In summary, we believe that this constitutes another immune evasion mechanism that bears striking similarities to that developed by cancer cells to evade immune surveillance.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico
| | - Elrashdy M. Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia;
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab 21934, Alexandria, Egypt
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
22
|
Mohammadi B, Dua K, Saghafi M, Singh SK, Heydarifard Z, Zandi M. COVID-19-induced autoimmune thyroiditis: Exploring molecular mechanisms. J Med Virol 2023; 95:e29001. [PMID: 37515444 DOI: 10.1002/jmv.29001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/30/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) damages multiple organs, including the thyroid, by direct invasion and cell entry via angiotensin-converting enzyme 2 or indirectly by promoting excessive inflammation in the body. The immune system is a critical factor in antiviral immunity and disease progression. In the context of SARS-CoV-2 infection, the immune system may become overly activated, resulting in a shift from regulatory to effector responses, which may subsequently promote the development and progression of autoimmune diseases. The incidence of autoimmune thyroid diseases, such as subacute thyroiditis, Graves' disease, and Hashimoto's thyroiditis, increases in individuals with COVID-19 infection. This phenomenon may be attributed to aberrant responses of T-cell subtypes, the presence of autoantibodies, impaired regulatory cell function, and excessive production of inflammatory cytokines, namely interleukin (IL)-6, IL-1β, interferon-γ, and tumor necrosis factor-α. Therefore, insights into the immune responses involved in the development of autoimmune thyroid disease according to COVID-19 can help identify potential therapeutic approaches and guide the development of effective interventions to alleviate patients' symptoms.
Collapse
Affiliation(s)
- Bita Mohammadi
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
- Innovated Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia
- Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohammadreza Saghafi
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
- Innovated Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Center in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Zahra Heydarifard
- Department of Virology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
- School of Medicine, Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Valdebenito S, Ono A, Rong L, Eugenin EA. The role of tunneling nanotubes during early stages of HIV infection and reactivation: implications in HIV cure. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:169-186. [PMID: 37476291 PMCID: PMC10355284 DOI: 10.1515/nipt-2022-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 07/22/2023]
Abstract
Tunneling nanotubes (TNTs), also called cytonemes or tumor microtubes, correspond to cellular processes that enable long-range communication. TNTs are plasma membrane extensions that form tubular processes that connect the cytoplasm of two or more cells. TNTs are mostly expressed during the early stages of development and poorly expressed in adulthood. However, in disease conditions such as stroke, cancer, and viral infections such as HIV, TNTs proliferate, but their role is poorly understood. TNTs function has been associated with signaling coordination, organelle sharing, and the transfer of infectious agents such as HIV. Here, we describe the critical role and function of TNTs during HIV infection and reactivation, as well as the use of TNTs for cure strategies.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Akira Ono
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| |
Collapse
|
24
|
Gottschalk CG, Peterson D, Armstrong J, Knox K, Roy A. Potential molecular mechanisms of chronic fatigue in long haul COVID and other viral diseases. Infect Agent Cancer 2023; 18:7. [PMID: 36750846 PMCID: PMC9902840 DOI: 10.1186/s13027-023-00485-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Historically, COVID-19 emerges as one of the most devastating diseases of humankind, which creates an unmanageable health crisis worldwide. Until now, this disease costs millions of lives and continues to paralyze human civilization's economy and social growth, leaving an enduring damage that will take an exceptionally long time to repair. While a majority of infected patients survive after mild to moderate reactions after two to six weeks, a growing population of patients suffers for months with severe and prolonged symptoms of fatigue, depression, and anxiety. These patients are no less than 10% of total COVID-19 infected individuals with distinctive chronic clinical symptomatology, collectively termed post-acute sequelae of COVID-19 (PASC) or more commonly long-haul COVID. Interestingly, Long-haul COVID and many debilitating viral diseases display a similar range of clinical symptoms of muscle fatigue, dizziness, depression, and chronic inflammation. In our current hypothesis-driven review article, we attempt to discuss the molecular mechanism of muscle fatigue in long-haul COVID, and other viral diseases as caused by HHV6, Powassan, Epstein-Barr virus (EBV), and HIV. We also discuss the pathological resemblance of virus-triggered muscle fatigue with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS).
Collapse
Affiliation(s)
- Carl Gunnar Gottschalk
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,grid.267468.90000 0001 0695 7223Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Daniel Peterson
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Jan Armstrong
- Simmaron Research INC, 948 Incline Way, Incline Village, NV 89451 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Konstance Knox
- grid.267468.90000 0001 0695 7223Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211 USA ,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI 53186 USA
| | - Avik Roy
- Simmaron Research INC, 948 Incline Way, Incline Village, NV, 89451, USA. .,Research and Development Laboratory, Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA. .,Coppe Laboratories, W229N1870 Westwood Dr, Waukesha, WI, 53186, USA.
| |
Collapse
|
25
|
Singh DD, Han I, Choi EH, Yadav DK. A Clinical Update on SARS-CoV-2: Pathology and Development of Potential Inhibitors. Curr Issues Mol Biol 2023; 45:400-433. [PMID: 36661514 PMCID: PMC9857284 DOI: 10.3390/cimb45010028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome) is highly infectious and causes severe acute respiratory distress syndrome (SARD), immune suppression, and multi-organ failure. For SARS-CoV-2, only supportive treatment options are available, such as oxygen supportive therapy, ventilator support, antibiotics for secondary infections, mineral and fluid treatment, and a significant subset of repurposed effective drugs. Viral targeted inhibitors are the most suitable molecules, such as ACE2 (angiotensin-converting enzyme-2) and RBD (receptor-binding domain) protein-based inhibitors, inhibitors of host proteases, inhibitors of viral proteases 3CLpro (3C-like proteinase) and PLpro (papain-like protease), inhibitors of replicative enzymes, inhibitors of viral attachment of SARS-CoV-2 to the ACE2 receptor and TMPRSS2 (transmembrane serine proteinase 2), inhibitors of HR1 (Heptad Repeat 1)-HR2 (Heptad Repeat 2) interaction at the S2 protein of the coronavirus, etc. Targeting the cathepsin L proteinase, peptide analogues, monoclonal antibodies, and protein chimaeras as RBD inhibitors interferes with the spike protein's ability to fuse to the membrane. Targeting the cathepsin L proteinase, peptide analogues, monoclonal antibodies, and protein chimaeras as RBD inhibitors interferes with the spike protein's ability to fuse to the membrane. Even with the tremendous progress made, creating effective drugs remains difficult. To develop COVID-19 treatment alternatives, clinical studies are examining a variety of therapy categories, including antibodies, antivirals, cell-based therapy, repurposed diagnostic medicines, and more. In this article, we discuss recent clinical updates on SARS-CoV-2 infection, clinical characteristics, diagnosis, immunopathology, the new emergence of variant, SARS-CoV-2, various approaches to drug development and treatment options. The development of therapies has been complicated by the global occurrence of many SARS-CoV-2 mutations. Discussion of this manuscript will provide new insight into drug pathophysiology and drug development.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, India
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
- Correspondence: (I.H.); (D.K.Y.); Tel.: +82-2-597-0365 (I.H. & D.K.Y.)
| | - Eun-Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Dharmendra Kumar Yadav
- Department of R&D Center, Arontier Co., Seoul 06735, Republic of Korea
- Correspondence: (I.H.); (D.K.Y.); Tel.: +82-2-597-0365 (I.H. & D.K.Y.)
| |
Collapse
|
26
|
Halfon P, Jordana S, Blachier S, Cartlamy P, Kbaier L, Psomas CK, Philibert P, Antoniotti G, Allemand-Sourrieu J, Rebaudet S, Cavaille G, Stavris C, Retornaz F, Chiche L, Penaranda G. Anti-spike protein to determine SARS-CoV-2 antibody levels: Is there a specific threshold conferring protection in immunocompromised patients? PLoS One 2023; 18:e0281257. [PMID: 37115758 PMCID: PMC10146437 DOI: 10.1371/journal.pone.0281257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/18/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Identifying a specific threshold level of SARS-CoV-2 antibodies that confers protection in immunocompromised patients has been very challenging. The aim was to assess the threshold of 264 binding antibody units (BAU)/ml using four different SARS-CoV-2 antibody assays (Abbott, Beckman, Roche, and Siemens) and to establish a new optimal threshold of protection for each of the four antibody assays. METHODS This study was performed on data retrieved from 69 individuals, who received at least one dose of the Pfizer/BioNTech BNT162b2 or Moderna COVID-19 vaccine (Spikevax) at the Alphabio Laboratory in Marseille, France (European Hospital, Alphabio-Biogroup). The results were compared to the percent inhibition calculated using a functional surrogate of a standardized virus neutralization test (Genscript). RESULTS Samples from 69 patients were analyzed. For a reference cutoff of 264 BAU/ml, assays showed moderate to good overall concordance with Genscript: 87% concordance for Abbott, 78% for Beckman, 75% for Roche, and 88% for Siemens. Overall concordance increased consistently after applying new thresholds, i.e., 148 BAU/ml (Abbott), 48 (Beckman), 559 (Roche), and 270 (Siemens). CONCLUSION We suggest specific adjusted thresholds (BAU/ml) for the four commercial antibody assays that are used to assess pre-exposure prophylaxis in immunocompromised patients.
Collapse
Affiliation(s)
- Philippe Halfon
- Laboratoire Alphabio-Biogroup, Marseille, France
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | | | | | | | | | - Christina K Psomas
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Patrick Philibert
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | | | - Julie Allemand-Sourrieu
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Stanislas Rebaudet
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Guilhem Cavaille
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Chloé Stavris
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Frédérique Retornaz
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | - Laurent Chiche
- Department of Infectious Diseases and Internal Medicine, Hôpital Européen, Marseille, France
| | | |
Collapse
|
27
|
Srivastava A, Hollenbach JA. The immunogenetics of COVID-19. Immunogenetics 2022; 75:309-320. [PMID: 36534127 PMCID: PMC9762652 DOI: 10.1007/s00251-022-01284-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/19/2022] [Indexed: 12/23/2022]
Abstract
The worldwide coronavirus disease 2019 pandemic was sparked by the severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2) that first surfaced in December 2019 (COVID-19). The effects of COVID-19 differ substantially not just between patients individually but also between populations with different ancestries. In humans, the human leukocyte antigen (HLA) system coordinates immune regulation. Since HLA molecules are a major component of antigen-presenting pathway, they play an important role in determining susceptibility to infectious disease. It is likely that differential susceptibility to SARS-CoV-2 infection and/or disease course in COVID-19 in different individuals could be influenced by the variations in the HLA genes which are associated with various immune responses to SARS-CoV-2. A growing number of studies have identified a connection between HLA variation and diverse COVID-19 outcomes. Here, we review research investigating the impact of HLA on individual responses to SARS-CoV-2 infection and/or progression, also discussing the significance of MHC-related immunological patterns and its use in vaccine design.
Collapse
Affiliation(s)
- Anshika Srivastava
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| | - Jill A. Hollenbach
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
28
|
Host Protective Immunity against Severe Acute Respiratory Coronavirus 2 (SARS-CoV-2) and the COVID-19 Vaccine-Induced Immunity against SARS-CoV-2 and Its Variants. Viruses 2022; 14:v14112541. [PMID: 36423150 PMCID: PMC9697230 DOI: 10.3390/v14112541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The world is now apparently at the last/recovery stage of the COVID-19 pandemic, starting from 29 December 2019, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the progression of time, several mutations have taken place in the original SARS-CoV-2 Wuhan strain, which have generated variants of concern (VOC). Therefore, combatting COVID-19 has required the development of COVID-19 vaccines using several platforms. The immunity induced by those vaccines is vital to study in order to assure total protection against SARS-CoV-2 and its emerging variants. Indeed, understanding and identifying COVID-19 protection mechanisms or the host immune responses are of significance in terms of designing both new and repurposed drugs as well as the development of novel vaccines with few to no side effects. Detecting the immune mechanisms for host protection against SARS-CoV-2 and its variants is crucial for the development of novel COVID-19 vaccines as well as to monitor the effectiveness of the currently used vaccines worldwide. Immune memory in terms of the production of neutralizing antibodies (NAbs) during reinfection is also very crucial to formulate the vaccine administration schedule/vaccine doses. The response of antigen-specific antibodies and NAbs as well as T cell responses, along with the protective cytokine production and the innate immunity generated upon COVID-19 vaccination, are discussed in the current review in comparison to the features of naturally induced protective immunity.
Collapse
|
29
|
Vinjamuri S, Li L, Bouvier M. SARS-CoV-2 ORF8: One protein, seemingly one structure, and many functions. Front Immunol 2022; 13:1035559. [PMID: 36353628 PMCID: PMC9637571 DOI: 10.3389/fimmu.2022.1035559] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/10/2022] [Indexed: 07/30/2023] Open
Abstract
SARS-CoV-2 is the virus responsible for the COVID-19 pandemic. The genome of SARS-CoV-2 encodes nine accessory proteins that are involved in host-pathogen interaction. ORF8 is unique among these accessory proteins. SARS-CoV-2 ORF8 shares a surprisingly low amino acid sequence similarity with SARS-COV ORF8 (30%), and it is presumed to have originated from bat. Studies have shown that ORF8 exerts multiple different functions that interfere with host immune responses, including the downregulation of MHC class I molecules. These functions may represent strategies of host immune evasion. The x-ray crystal structure of ORF8 revealed an immunoglobulin-like domain with several distinguishing features. To date, there are numerous unanswered questions about SARS-CoV-2 ORF8 protein and its structure-function relationship that we discuss in this mini-review. A better understanding of how ORF8 interacts with components of the immune system is needed for elucidating COVID-19 pathogenesis and to develop new avenues for the treatment of the disease.
Collapse
Affiliation(s)
| | | | - Marlene Bouvier
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL, United States
| |
Collapse
|
30
|
Roggero PF, Calistri A, Palù G. On the intrinsic nature of viral pathogenesis: The assumption of a Darwinian paradigm to describe COVID-19 pandemic. Comput Struct Biotechnol J 2022; 20:5870-5872. [PMID: 36320938 PMCID: PMC9613777 DOI: 10.1016/j.csbj.2022.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/17/2022] [Accepted: 10/23/2022] [Indexed: 11/30/2022] Open
Abstract
Our hypothesis about evolution of the COVID-19 pandemic foresees an inverse relation between infectivity (R0) and lethality (L) of SARS-CoV-2. The above parameters are driven by a continuing mutation process granting the virus a clear survival advantage over virulence. For interpreting this relation we adopted a simple equation, R0 × L ≈ k, by which R0 and L depend upon a constant k, that corresponds to an intrinsic property of the viral species involved. The hypothesis was verified by following changes of the R0 and L terms of the formula in the different variants of SARS-CoV-2 that progressively appeared. A further validation came when the equation was applied to pandemic and epidemic influenza type A viruses, Ebola virus and measles virus. We believe this equation that considers virus biology in Darwinian terms could be extremely useful to better face infectious viral threats and validate virus-host molecular interactions relevant to viral pathogenesis.
Collapse
Affiliation(s)
- Pier Francesco Roggero
- Department of Molecular Medicine, University of Padua, via A. Gabelli 63, 35121 Padua, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, via A. Gabelli 63, 35121 Padua, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, via A. Gabelli 63, 35121 Padua, Italy,Italian Medicines Agency, Via del Tritone 181, 00187 Rome, Italy,Corresponding author at: Department of Department of Molecular Medicine, University of Padua, via A. Gabelli 63, 35121 Padua, Italy; Italian Medicines Agency, Via del Tritone 181, 00187, Rome, Italy
| |
Collapse
|