1
|
Srivastava S, Dhoundiyal S, Kumar S, Kaur A, Khatib MN, Gaidhane S, Zahiruddin QS, Mohanty A, Henao-Martinez AF, Krsak M, Rodriguez-Morales AJ, Montenegro-Idrogo JJ, Bonilla-Aldana DK, Sah R. Yellow Fever: Global Impact, Epidemiology, Pathogenesis, and Integrated Prevention Approaches. LE INFEZIONI IN MEDICINA 2024; 32:434-450. [PMID: 39660161 PMCID: PMC11627485 DOI: 10.53854/liim-3204-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/10/2024] [Indexed: 12/12/2024]
Abstract
Yellow fever poses a substantial global health concern as one of the re-emerging diseases with pandemic potential in a scenario of the worldwide distribution of some vectors (such as Aedes aegypti); in the context of climatic change, an unclear knowledge about the immune behaviour of the virus, between other determinants. This review details the historical foundations, intricate evolution of geographical spread, and transmission mechanisms of the disease to understand the behaviour of outbreaks over time in a multifactorial context that could be difficult to understand. This article approaches to epidemiological, pathophysiological, immunological, social determinants, and climatic crisis by understanding possible control mechanisms and anticipating potential future epidemics. This article explores the evidence of yellow fever virus (YFV) pathogenesis and its complex interactions with the immune response in the host, the vector, and in the context of immunisation. These discussions contribute to a more comprehensive understanding of the disease's progression. Despite the global presence of the vector and other factors that could facilitate an epidemic spread, yellow fever outbreaks have remained confined to specific endemic areas. This limited distribution is not entirely understood. However, it may be influenced by the complex immune interactions between the virus, the vector, and the host, preventing its spread to other regions.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017,
India
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201,
India
| | - Shivang Dhoundiyal
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, 333031 Rajasthan,
India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi 110017,
India
| | - Awaneet Kaur
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201,
India
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha,
India
| | - Shilpa Gaidhane
- One Health Centre (COHERD), Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha,
India
| | - Quazi Syed Zahiruddin
- Global Health Academy, Division of Evidence Synthesis, School of Epidemiology and Public Health and Research, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha.
India
| | - Aroop Mohanty
- Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur,
India
| | - Andres F. Henao-Martinez
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO,
USA
| | - Martin Krsak
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO,
USA
| | - Alfonso J. Rodriguez-Morales
- Master of Clinical Epidemiology and Biostatistics Program, Faculty of Health Sciences, Universidad Científica del Sur, Lima,
Peru
- Grupo de Investigación Biomedicina, Facultad de Medicina, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Pereira 660003, Risaralda,
Colombia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut,
Lebanon
| | - Juan J. Montenegro-Idrogo
- Master of Clinical Epidemiology and Biostatistics Program, Faculty of Health Sciences, Universidad Científica del Sur, Lima,
Peru
- Infectious and Tropical Diseases Service, Hospital Nacional Dos de Mayo, Lima,
Peru
| | | | - Ranjit Sah
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune 411018, Maharashtra,
India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra,
India
- SR Sanjeevani Hospital, Kalyanpur-10, Siraha,
Nepal
| |
Collapse
|
2
|
Muthukutty P, MacDonald J, Yoo SY. Combating Emerging Respiratory Viruses: Lessons and Future Antiviral Strategies. Vaccines (Basel) 2024; 12:1220. [PMID: 39591123 PMCID: PMC11598775 DOI: 10.3390/vaccines12111220] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging viral diseases, including seasonal illnesses and pandemics, pose significant global public health risks. Respiratory viruses, particularly coronaviruses and influenza viruses, are associated with high morbidity and mortality, imposing substantial socioeconomic burdens. This review focuses on the current landscape of respiratory viruses, particularly influenza and SARS-CoV-2, and their antiviral treatments. It also discusses the potential for pandemics and the development of new antiviral vaccines and therapies, drawing lessons from past outbreaks to inform future strategies for managing viral threats.
Collapse
Affiliation(s)
| | | | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea; (P.M.); (J.M.)
| |
Collapse
|
3
|
Alqassim AY, Badedi M, Muaddi MA, Alharbi AA, Jareebi MA, Makeen AM, El-Setouhy M, Albasheer OB, Sabai A, Sahly A. Shifting spatial, temporal and demographic patterns of dengue incidence and associated meteorological factors in Jazan Region of Saudi Arabia from 2015-2020. J Vector Borne Dis 2024; 61:444-451. [PMID: 38634364 DOI: 10.4103/jvbd.jvbd_15_24] [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: 01/28/2024] [Accepted: 04/16/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND OBJECTIVES Dengue poses a considerable public health threat in Saudi Arabia, with escalating outbreaks in Jazan, where seasonal rains create ideal mosquito breeding conditions. Elucidating local epidemiological dynamics is imperative to strengthen evidence-based prevention policies. This study analyzed the spatiotemporal, demographic, and meteorological patterns of dengue in Jazan from 2015-2020. METHODS This retrospective cross-sectional study utilized surveillance records for 3427 confirmed dengue cases. Descriptive analyses characterized geographic, seasonal, age, gender, and nationality distributions. Forecasting models project expected epidemics through 2025. Regression analysis identified climate factors associated with monthly case counts. RESULTS Dengue exhibited shifting seasonal peaks, transitioning into year-round transmission by 2019, indicating endemic establishment. Cases clustered in different high-burden sectors annually, requiring localized vector control. The majority of affected individuals were young male adults, with gender gaps narrowing over time. Saudi nationals had an escalating incidence, but non-citizens showed a higher risk, signaling importation threats. Seasonal outbreaks were associated with temperature, wind speed, and direction. INTERPRETATION CONCLUSION Enhanced surveillance, outbreak forecasting, targeted control activities, and integrated prevention policies grounded in continuous evidence assessment can effectively address endemic dengue transmission in Jazan. This study provides key insights to optimize data-driven decision-making for dengue control in Saudi Arabia.
Collapse
Affiliation(s)
- Ahmad Y Alqassim
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Mohammed Badedi
- Administration of Research and Studies, Jazan Health Department, Jazan City, Jazan, Saudi Arabia
| | - Mohammed A Muaddi
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Abdullah A Alharbi
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Mohammad A Jareebi
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Anwar M Makeen
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Maged El-Setouhy
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Osama B Albasheer
- Family and Community Medicine Department, Faculty of Medicine, Jazan University, Jazan City, Jazan, Saudi Arabia
| | - Abdullah Sabai
- Directorate of Primary Health Care Centers, Jazan Health Department, Jazan City, Jazan, Saudi Arabia
| | - Ahmed Sahly
- Public Health Administration, Jazan Health Department, Jazan City, Jazan, Saudi Arabia
| |
Collapse
|
4
|
Kumar DS, Prasanth K, Bhandari A, Kumar Jha V, Naveen A, Prasanna M. Innovations and Challenges in the Development of COVID-19 Vaccines for a Safer Tomorrow. Cureus 2024; 16:e60015. [PMID: 38854201 PMCID: PMC11162516 DOI: 10.7759/cureus.60015] [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] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Vaccination, a historically effective public health intervention, has shielded millions from various diseases. Lessons from severe acute respiratory syndrome coronavirus (SARS-CoV) have improved COVID-19 vaccine development. Despite mRNA vaccines' efficacy, emerging variants pose challenges, exhibiting increased transmissibility, infectivity, and severity. Developing COVID-19 vaccines has faced hurdles due to urgency, limited virus understanding, and the need for safe solutions. Genetic variability necessitates continuous vaccine adjustments and production challenges demand scaling up manufacturing with stringent quality control. This review explores SARS-CoV-2's evolution, upcoming mutations that challenge vaccines, and strategies such as structure-based, T cell-based, respiratory mucosal-based, and nanotechnology approaches for vaccine development. This review insight provides a roadmap for navigating virus evolution and improving vaccine development.
Collapse
Affiliation(s)
- Devika S Kumar
- Research, Panimalar Medical College Hospital and Research Institute, Chennai, IND
| | - Krishna Prasanth
- Department of Community Medicine, Sree Balaji Medical College and Hospital, Chennai, IND
| | - Ashni Bhandari
- Department of Community Medicine, Sree Balaji Medical College and Hospital, Chennai, IND
| | - Vivek Kumar Jha
- Department of Audiology and Speech Language Pathology, Shree Guru Gobind Singh Tricentenary (SGT) University, Haryana, IND
| | - Avula Naveen
- Pharmacology and Therapeutics, All India Institute Of Medical Science Bilaspur, Bilaspur, IND
| | - Muthu Prasanna
- Pharmaceutics, Pharmaceutical Biotechnology, Surya School of Pharmacy, Surya Group of Institutions, Villupuram, IND
| |
Collapse
|
5
|
Khan RU, Kumar R, Haq AU, Khan I, Shabaz M, Khan F. Blockchain-Based Trusted Tracking Smart Sensing Network to Prevent the Spread of Infectious Diseases. Ing Rech Biomed 2024; 45:100829. [DOI: 10.1016/j.irbm.2024.100829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
|
6
|
Arshad SF, Rehana R, Saleem MA, Usman M, Arshad HJ, Rizwana R, Shakeela S, Rukh AS, Khan IA, Hayssam MA, Anwar M. Multi-epitopes vaccine design for surface glycoprotein against SARS-CoV-2 using immunoinformatic approach. Heliyon 2024; 10:e24186. [PMID: 38298616 PMCID: PMC10827691 DOI: 10.1016/j.heliyon.2024.e24186] [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: 10/24/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Background The recent COVID vaccinations have successfully reduced death and severity but did not stop the transmission of viruses by the emerging SARS-CoV-2 strain. There is a need for better and long-lasting dynamic vaccines for numerous prevailing strains and the evolving SARS-CoV-2 virus, necessitating the development of broad-spectrum strains being used to stop infection by reducing the spread rate and re-infection. The spike (S) glycoprotein is one of the proteins expressed commonly in the early phases of SARS-CoV-2 infection. It has been identified as the most immunogenic protein of SARS-CoV-2. Methods In this study, advanced bioinformatics techniques have been exploited to design the novel multi-epitope vaccine using conserved S protein portions from widespread strains of SARS-CoV-2 to predict B cell and T cell epitopes. These epitopes were selected based on toxicity, antigenicity score and immunogenicity. Epitope combinations were used to construct the maximum potent multi-epitope construct with potential immunogenic features. EAAAK, AAY, and GPGPG were used as linkers to construct epitopes. Results The developed vaccine has shown positive results. After the chimeric vaccine construct was cloned into the PET28a (+) vector for expression screening in Escherichia coli, the potential expression of the construct was identified. Conclusion The construct vaccine performed well in computer-based immune response simulation and covered a variety of allelic populations. These computational results are more helpful for further analysis of our contract vaccine, which can finally help control and prevent SARS-CoV-2 infections worldwide.
Collapse
Affiliation(s)
- Sarmad Frogh Arshad
- Department of Biochemistry and Biotechnology, Muhammad Nawaz Shareef University of Agriculture, Multan, 66000, Pakistan
| | - Rehana Rehana
- Institute of Plant Breeding & Biotechnology (IPBB), Muhammad Nawaz Shareef University of Agriculture, Multan, 66000, Pakistan
| | - Muhammad Asif Saleem
- Department of Plant Breeding and Genetics, Bahauddin Zakaria University, Multan, 60800, Pakistan
| | - Muhammad Usman
- Department of Biochemistry and Biotechnology, Muhammad Nawaz Shareef University of Agriculture, Multan, 66000, Pakistan
| | - Hasan Junaid Arshad
- Centre of Agricultural Biochemistry and Biotechnology, University of Agriculture, Faisalabad, 38000, Pakistan
| | - Rizwana Rizwana
- Department of Biochemistry, Bahauddin Zakaria University, Multan, 60800, Pakistan
| | | | - Asma Shah Rukh
- Department of Pharmacy, College of Pharmacy Punjab University, Lahore, 54590, Pakistan
| | - Imran Ahmad Khan
- Department of Pharmacy, MNS University of Agriculture, Multan, 54590, Pakistan
| | - M. Ali Hayssam
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 1145, Saudi Arabia
| | - Muhammad Anwar
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, PR China
| |
Collapse
|
7
|
Chavda V, Yadav D, Parmar H, Brahmbhatt R, Patel B, Madhwani K, Jain M, Song M, Patel S. A Narrative Overview of Coronavirus Infection: Clinical Signs and Symptoms, Viral Entry and Replication, Treatment Modalities, and Management. Curr Top Med Chem 2024; 24:1883-1916. [PMID: 38859776 DOI: 10.2174/0115680266296095240529114058] [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: 01/01/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 06/12/2024]
Abstract
The global pandemic known as coronavirus disease (COVID-19) is causing morbidity and mortality on a daily basis. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV- -2) virus has been around since December 2019 and has infected a high number of patients due to its idiopathic pathophysiology and rapid transmission. COVID-19 is now deemed a newly identified "syndrome" condition since it causes a variety of unpleasant symptoms and systemic side effects following the pandemic. Simultaneously, it always becomes potentially hazardous when new variants develop during evolution. Its random viral etiology prevents accurate and suitable therapy. Despite the fact that multiple preclinical and research studies have been conducted to combat this lethal virus, and various therapeutic targets have been identified, the precise course of therapy remains uncertain. However, just a few drugs have shown efficacy in treating this viral infection in its early stages. Currently, several medicines and vaccinations have been licensed following clinical trial research, and many countries are competing to find the most potent and effective immunizations against this highly transmissible illness. For this narrative review, we used PubMed, Google Scholar, and Scopus to obtain epidemiological data, pre-clinical and clinical trial outcomes, and recent therapeutic alternatives for treating COVID-19 viral infection. In this study, we discussed the disease's origin, etiology, transmission, current advances in clinical diagnostic technologies, different new therapeutic targets, pathophysiology, and future therapy options for this devastating virus. Finally, this review delves further into the hype surrounding the SARS-CoV-2 illness, as well as present and potential COVID-19 therapies.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pathology, Stanford School of Medicine, Stanford University Medical Center, Palo Alto94305, CA, USA
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, South Korea
| | - Harisinh Parmar
- Department of Neurosurgery, Krishna institute of medical sciences, Karad, Maharashtra, India
| | - Raxit Brahmbhatt
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Bipin Patel
- Department of Medicine, Multispeciality, Trauma and ICCU Center, Sardar Hospital, Ahmedabad, 382352, Gujarat, India
| | - Kajal Madhwani
- Department of Life Science, University of Westminster, London, W1B 2HW, United Kingdom
| | - Meenu Jain
- Gajra Raja Medical College, Gwalior, 474009, Madhya Pradesh, India
| | - Minseok Song
- Department of Life Science, Yeungnam University, South Korea
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmedabad, 382481, Gujarat, India
| |
Collapse
|
8
|
He Y, Hu Q, San S, Kasputis T, Splinter MGD, Yin K, Chen J. CRISPR-based Biosensors for Human Health: A Novel Strategy to Detect Emerging Infectious Diseases. Trends Analyt Chem 2023; 168:117342. [PMID: 37840598 PMCID: PMC10571337 DOI: 10.1016/j.trac.2023.117342] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Infectious diseases (such as sepsis, influenza, and malaria), caused by various pathogenic bacteria and viruses, are widespread across the world. Early and rapid detection of disease-related pathogens is necessary to reduce their spread in the world and prevent their potential global pandemics. The clustered regularly interspaced short palindromic repeats (CRISPR) technology, as the next-generation molecular diagnosis technique, holds immense promise in the detection of infectious diseases because of its remarkable advantages, including supreme flexibility, sensitivity, and specificity. While numerous CRISPR-based biosensors have been developed for application in environmental monitoring, food safety, and point-of-care diagnosis, there remains a critical need to summarize and explore their potential in human health. This review aims to address this gap by focusing on the latest advancements in CRISPR-based biosensors for infectious disease detection. We provide an overview of the current status, pre-amplification methods, the unique feature of each CRISPR system, and the design of CRISPR-based biosensing strategies to detect disease-associated nucleic acids. Last but not least, the review analyzes the current challenges and provides future perspectives, which will contribute to developing more effective CRISPR-based biosensors for human health.
Collapse
Affiliation(s)
- Yawen He
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Qinqin Hu
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, People’s Republic of China
| | - Samantha San
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Tom Kasputis
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Kun Yin
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, People’s Republic of China
| | - Juhong Chen
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
9
|
Carlini F, Lusi V, Rizzi C, Assogna F, Laroni A. Cladribine Tablets Mode of Action, Learning from the Pandemic: A Narrative Review. Neurol Ther 2023; 12:1477-1490. [PMID: 37421556 PMCID: PMC10444742 DOI: 10.1007/s40120-023-00520-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/16/2023] [Indexed: 07/10/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system, characterized by chronic, inflammatory, demyelinating, and neurodegenerative processes. MS management relies on disease-modifying drugs that suppress/modulate the immune system. Cladribine tablets (CladT) have been approved by different health authorities for patients with various forms of relapsing MS. The drug has been demonstrated to deplete CD4+ and CD8+ T-cells, with a higher effect described in the former, and to decrease total CD19+, CD20+, and naive B-cell counts. COVID-19 is expected to become endemic, suggesting its potential infection risk for immuno-compromised patients, including MS patients treated with disease-modifying drugs. We report here the available data on disease-modifying drug-treated-MS patients and COVID-19 infection and vaccination, with a focus on CladT. MS patients treated with CladT are not at higher risk of developing severe COVID-19. While anti-SARS-CoV-2 vaccination is recommended in all MS patients with guidelines addressing vaccination timing according to the different disease-modifying drugs, no vaccination timing restrictions seem to be necessary for cladribine, based on its mechanism of action and available evidence. Published data suggest that CladT treatment does not impact the production of anti-SARS-CoV-2 antibodies after COVID-19 vaccination, possibly due to its relative sparing effect on naïve B-cells and the rapid B-cell reconstitution following treatment. Slightly lower specific T-cell responses are likely not impacting the risk of breakthrough COVID-19. It could be stated that cladribine's transient effect on innate immune cells likely contributes to maintaining an adequate first line of defense against the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Federico Carlini
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa, Italy
| | - Valeria Lusi
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa, Italy
| | - Caterina Rizzi
- Merck Serono S.P.A., Italy an Affiliate of Merck KGaA, Piazza del Pigneto 9, Rome, Italy
| | - Francesco Assogna
- Merck Serono S.P.A., Italy an Affiliate of Merck KGaA, Piazza del Pigneto 9, Rome, Italy
| | - Alice Laroni
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genoa, Italy.
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Largo Daneo 3, Genoa, Italy.
| |
Collapse
|
10
|
Detection of live SARS-CoV-2 virus and its variants by specially designed SERS-active substrates and spectroscopic analyses. Anal Chim Acta 2023; 1256:341151. [PMID: 37037632 PMCID: PMC10060322 DOI: 10.1016/j.aca.2023.341151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/07/2023] [Accepted: 03/26/2023] [Indexed: 04/05/2023]
Abstract
A method using label-free surface enhanced Raman spectroscopy (SERS) based on substrate design is provided for an early detection and differentiation of spike glycoprotein mutation sites in live SARS-CoV-2 variants. Two SERS-active substrates, Au nanocavities (Au NCs) and Au NPs on porous ZrO2 (Au NPs/pZrO2), were used to identify specific peaks of A.3, Alpha, and Delta variants at different concentrations and demonstrated the ability to provide their SERS spectra with detection limits of 0.1–1.0% (or 104−5 copies/mL). Variant identification can be achieved by cross-examining reference spectra and analyzing the substrate-analyte relationship between the suitability of the analyte upon the hotspot(s) formed at high concentrations and the effective detection distance at low concentrations. Mutation sites on the S1 chain of the spike glycoprotein for each variant may be related and distinguishable. This method does not require sample preprocessing and therefore allows for fast screening, which is of high value for more comprehensive and specific studies to distinguish upcoming variants.
Collapse
|
11
|
Kakavandi S, Zare I, VaezJalali M, Dadashi M, Azarian M, Akbari A, Ramezani Farani M, Zalpoor H, Hajikhani B. Structural and non-structural proteins in SARS-CoV-2: potential aspects to COVID-19 treatment or prevention of progression of related diseases. Cell Commun Signal 2023; 21:110. [PMID: 37189112 PMCID: PMC10183699 DOI: 10.1186/s12964-023-01104-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/15/2023] [Indexed: 05/17/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a new member of the Coronaviridae family known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There are structural and non-structural proteins (NSPs) in the genome of this virus. S, M, H, and E proteins are structural proteins, and NSPs include accessory and replicase proteins. The structural and NSP components of SARS-CoV-2 play an important role in its infectivity, and some of them may be important in the pathogenesis of chronic diseases, including cancer, coagulation disorders, neurodegenerative disorders, and cardiovascular diseases. The SARS-CoV-2 proteins interact with targets such as angiotensin-converting enzyme 2 (ACE2) receptor. In addition, SARS-CoV-2 can stimulate pathological intracellular signaling pathways by triggering transcription factor hypoxia-inducible factor-1 (HIF-1), neuropilin-1 (NRP-1), CD147, and Eph receptors, which play important roles in the progression of neurodegenerative diseases like Alzheimer's disease, epilepsy, and multiple sclerosis, and multiple cancers such as glioblastoma, lung malignancies, and leukemias. Several compounds such as polyphenols, doxazosin, baricitinib, and ruxolitinib could inhibit these interactions. It has been demonstrated that the SARS-CoV-2 spike protein has a stronger affinity for human ACE2 than the spike protein of SARS-CoV, leading the current study to hypothesize that the newly produced variant Omicron receptor-binding domain (RBD) binds to human ACE2 more strongly than the primary strain. SARS and Middle East respiratory syndrome (MERS) viruses against structural and NSPs have become resistant to previous vaccines. Therefore, the review of recent studies and the performance of current vaccines and their effects on COVID-19 and related diseases has become a vital need to deal with the current conditions. This review examines the potential role of these SARS-CoV-2 proteins in the initiation of chronic diseases, and it is anticipated that these proteins could serve as components of an effective vaccine or treatment for COVID-19 and related diseases. Video Abstract.
Collapse
Affiliation(s)
- Sareh Kakavandi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Maryam VaezJalali
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Dadashi
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Azarian
- Department of Radiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Abdullatif Akbari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Pal T, Anand U, Sikdar Mitra S, Biswas P, Tripathi V, Proćków J, Dey A, Pérez de la Lastra JM. Harnessing and bioprospecting botanical-based herbal medicines against potential drug targets for COVID-19: a review coupled molecular docking studies. J Biomol Struct Dyn 2023:1-23. [PMID: 37105230 DOI: 10.1080/07391102.2023.2187634] [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: 04/29/2023]
Abstract
Since the end of February 2020, the world has come to a standstill due to the virus SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2). Since then, the global scientific community has explored various remedies and treatments against this virus, including natural products that have always been a choice because of their many benefits. Various known phytochemicals are well documented for their antiviral properties. Research is being carried out to discover new natural plant products or existing ones as a treatment measure for this disease. The three important targets in this regard are-papain like protease (PLpro), spike protein, and 3 chymotrypsin like proteases (3CLpro). Various docking studies are also being elucidated to identify the phytochemicals that modulate crucial proteins of the virus. The paper is simultaneously a comprehensive review that covers recent advances in the domain of the effect of various botanically derived natural products as an alternative treatment approach against Coronavirus Disease 2019 (COVID-19). Furthermore, the docking analyses revealed that rutin (inhibitor of the major protease of SARS-CoV-2), gallocatechin (e.g., interacting with 03 hydrogen bonds with a spike-like protein), lycorine (showing the best binding affinity with amino acids GLN498, THR500 and GLY446 of the spike-like protein), and quercetrin (inhabiting at its residues ASP216, PHE219, and ILE259) are promising inhibitors of SARS‑CoV‑2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Tarun Pal
- Zuckerberg Institute for Water Research, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion, Israel
| | | | - Shreya Sikdar Mitra
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Protha Biswas
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - Vijay Tripathi
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, Uttar Pradesh, India
| | - Jarosław Proćków
- Department of Plant Biology, Institute of Environmental Biology, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules Research Group, Instituto de Productos Naturales y Agrobiología, IPNA-CSIC, Tenerife, Spain
| |
Collapse
|
13
|
Kubra KT, Ali R. Modeling and analysis of novel COVID-19 outbreak under fractal-fractional derivative in Caputo sense with power-law: a case study of Pakistan. MODELING EARTH SYSTEMS AND ENVIRONMENT 2023; 9:1-18. [PMID: 37361699 PMCID: PMC10019432 DOI: 10.1007/s40808-023-01747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/11/2023] [Indexed: 03/18/2023]
Abstract
In this paper, a five-compartment model is used to explore the dynamics of the COVID-19 pandemic, taking the vaccination campaign into account. The present model consists of five components that lead to a system of five ordinary differential equations. In this paper, we examined the disease from the perspective of a fractal fractional derivative in the Caputo sense with a power law type kernal. The model is also fitted with real data for Pakistan between June 1, 2020, and March 8, 2021. The fundamental mathematical characteristics of the model have been investigated thoroughly. We have calculated the equilibrium points and the reproduction number for the model and obtained the feasible region for the system. The existence and stability criteria of the model have been validated using the Banach fixed point theory and the Picard successive approximation technique. Furthermore, we have conducted stability analysis for both the disease-free and endemic equilibrium states. On the basis of sensitivity analysis and the dynamics of the threshold parameter, we have estimated the effectiveness of vaccination and identified potential control strategies for the disease using the proposed model outbreaks. The stability of the concerned solution in Ulam-Hyers and Ulam-Hyers-Rassias sense is also investigated. For the proposed problem, some results regarding basic reproduction numbers and stability analysis for various parameters are represented graphically. Matlab software is used for numerical illustrations. Graphical representations are given for different fractional orders and for various parametric values.
Collapse
Affiliation(s)
- Khadija Tul Kubra
- Department of Mathematics, Government College University Faisalabad, Faisalabad, 38000 Pakistan
| | - Rooh Ali
- Department of Mathematics, Government College University Faisalabad, Faisalabad, 38000 Pakistan
| |
Collapse
|
14
|
Park J, Champion JA. Effect of Antigen Structure in Subunit Vaccine Nanoparticles on Humoral Immune Responses. ACS Biomater Sci Eng 2023; 9:1296-1306. [PMID: 36848229 PMCID: PMC10015428 DOI: 10.1021/acsbiomaterials.2c01516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023]
Abstract
Subunit vaccines offer numerous attractive features, including good safety profiles and well-defined components with highly characterized properties because they do not contain whole pathogens. However, vaccine platforms based on one or few selected antigens are often poorly immunogenic. Several advances have been made in improving the effectiveness of subunit vaccines, including nanoparticle formulation and/or co-administration with adjuvants. Desolvation of antigens into nanoparticles is one approach that has been successful in eliciting protective immune responses. Despite this advance, damage to the antigen structure by desolvation can compromise the recognition of conformational antigens by B cells and the subsequent humoral response. Here, we used ovalbumin as a model antigen to demonstrate enhanced efficacy of subunit vaccines by preserving antigen structures in nanoparticles. An altered antigen structure due to desolvation was first validated by GROMACS and circular dichroism. Desolvant-free nanoparticles with a stable ovalbumin structure were successfully synthesized by directly cross-linking ovalbumin or using ammonium sulfate to form nanoclusters. Alternatively, desolvated OVA nanoparticles were coated with a layer of OVA after desolvation. Vaccination with salt-precipitated nanoparticles increased OVA-specific IgG titers 4.2- and 22-fold compared to the desolvated and coated nanoparticles, respectively. In addition, enhanced affinity maturation by both salt precipitated and coated nanoparticles was displayed in contrast to desolvated nanoparticles. These results demonstrate both that salt-precipitated antigen nanoparticles are a potential new vaccine platform with significantly improved humoral immunity and a functional value of preserving antigen structures in vaccine nanoparticle design.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
15
|
Soeroto AY, Yanto TA, Kurniawan A, Hariyanto TI. Efficacy and safety of tixagevimab-cilgavimab as pre-exposure prophylaxis for COVID-19: A systematic review and meta-analysis. Rev Med Virol 2023; 33:e2420. [PMID: 36617704 DOI: 10.1002/rmv.2420] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
Some proportions of populations, such as immunocompromised patients and organ transplant recipients might have inadequate immune responses to the vaccine for coronavirus disease 2019 (COVID-19). For these groups of populations, administering monoclonal antibodies might offer some additional protection. This review sought to analyze the effectiveness and safety of tixagevimab-cilgavimab (Evusheld) as pre-exposure prophylaxis against COVID-19. We used specific keywords to comprehensively search for potential studies on PubMed, Scopus, Europe PMC, and ClinicalTrials.gov sources until 3 September 2022. We collected all published articles that analyzed tixagevimab-cilgavimab on the course of COVID-19. Review Manager 5.4 was utilized for statistical analysis. Six studies were included. Our pooled analysis revealed that tixagevimab-cilgavimab prophylaxis may decrease the rate of SARS-CoV-2 infection (OR: 0.24; 95% CI: 0.15-0.40, p < 0.00001, I2 = 75%), lower COVID-19 hospitalization rate (OR: 0.13; 95% CI: 0.07-0.24, p < 0.00001, I2 = 0%), decrease the severity risk (OR: 0.13; 95% CI: 0.07-0.24, p < 0.00001, I2 = 0%), and lower COVID-19 deaths (OR: 0.17; 95% CI: 0.03-0.99, p = 0.05, I2 = 72%). In the included studies, no major adverse events were reported. This study proposes that tixagevimab-cilgavimab was effective and safe for preventing COVID-19. Tixagevimab-cilgavimab may be offered to those who cannot be vaccinated or have inadequate immune response from the COVID-19 vaccine to give additional protection.
Collapse
Affiliation(s)
- Arto Yuwono Soeroto
- Department of Internal Medicine, Division of Pulmonology and Critical Illness, Padjadjaran University, Bandung, West Java, Indonesia
| | - Theo Audi Yanto
- Department of Internal Medicine, Faculty of Medicine, Pelita Harapan University, Tangerang, Indonesia
| | - Andree Kurniawan
- Department of Internal Medicine, Faculty of Medicine, Pelita Harapan University, Tangerang, Indonesia
| | | |
Collapse
|
16
|
Bayani F, Hashkavaei NS, Arjmand S, Rezaei S, Uskoković V, Alijanianzadeh M, Uversky VN, Ranaei Siadat SO, Mozaffari-Jovin S, Sefidbakht Y. An overview of the vaccine platforms to combat COVID-19 with a focus on the subunit vaccines. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 178:32-49. [PMID: 36801471 PMCID: PMC9938630 DOI: 10.1016/j.pbiomolbio.2023.02.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging virus that has caused the recent coronavirus disease (COVID-19) global pandemic. The current approved COVID-19 vaccines have shown considerable efficiency against hospitalization and death. However, the continuation of the pandemic for more than two years and the likelihood of new strain emergence despite the global rollout of vaccination highlight the immediate need for the development and improvement of vaccines. mRNA, viral vector, and inactivated virus vaccine platforms were the first members of the worldwide approved vaccine list. Subunit vaccines. which are vaccines based on synthetic peptides or recombinant proteins, have been used in lower numbers and limited countries. The unavoidable advantages of this platform, including safety and precise immune targeting, make it a promising vaccine with wider global use in the near future. This review article summarizes the current knowledge on different vaccine platforms, focusing on the subunit vaccines and their clinical trial advancements against COVID-19.
Collapse
Affiliation(s)
- Fatemeh Bayani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | | | - Sareh Arjmand
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Shokouh Rezaei
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Vuk Uskoković
- Department of Mechanical Engineering, San Diego State University, San Diego, CA, 92182, USA; TardigradeNano LLC, Irvine, CA, 92604, USA
| | - Mahdi Alijanianzadeh
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| | | | - Sina Mozaffari-Jovin
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yahya Sefidbakht
- Protein Research Center, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
17
|
Zhao M, Slotkin R, Sheth AH, Pischel L, Kyriakides TC, Emu B, McNamara C, Shi Q, Delgobbo J, Xu J, Marhoffer E, Mercer-Falkoff A, Holleck J, Ardito D, Sutton RE, Gupta S. Serum Neutralizing Antibody Titers 12 Months After Coronavirus Disease 2019 Messenger RNA Vaccination: Correlation to Clinical Variables in an Adult, US Population. Clin Infect Dis 2023; 76:e391-e399. [PMID: 35639598 PMCID: PMC9278145 DOI: 10.1093/cid/ciac416] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We studied whether comorbid conditions affect strength and duration of immune responses after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) messenger RNA vaccination in a US-based, adult population. METHODS Sera (before and after BNT162b2 vaccination) were tested serially up to 12 months after 2 doses of vaccine for SARS-CoV-2-anti-Spike neutralizing capacity by pseudotyping assay in 124 individuals; neutralizing titers were correlated to clinical variables with multivariate regression. Postbooster (third dose) effect was measured at 1 and 3 months in 72 and 88 subjects, respectively. RESULTS After completion of primary vaccine series, neutralizing antibody half maximal inhibitory concentration (IC50) values were high at 1 month (14-fold increase from prevaccination), declined at 6 months (3.3-fold increase), and increased at 1 month postbooster (41.5-fold increase). Three months postbooster, IC50 decreased in coronavirus disease (COVID)-naïve individuals (18-fold increase) and increased in prior COVID 2019 (COVID-19+) individuals (132-fold increase). Age >65 years (β = -0.94, P = .001) and malignancy (β = -0.88, P = .002) reduced strength of response at 1 month. Both neutralization strength and durability at 6 months, respectively, were negatively affected by end-stage renal disease ([β = -1.10, P = .004]; [β = -0.66, P = .014]), diabetes mellitus ([β = -0.57, P = .032]; [β = -0.44, P = .028]), and systemic steroid use ([β = -0.066, P = .032]; [β = -0.55, P = .037]). Postbooster IC50 was robust against WA-1 and B.1.617.2. Postbooster neutralization increased with prior COVID-19 (β = 2.9, P < .0001), and malignancy reduced neutralization response (β = -0.68, P = .03), regardless of infection status. CONCLUSIONS Multiple clinical factors affect the strength and duration of neutralization response after primary series vaccination, but not the postbooster dose strength. Malignancy was associated with lower booster-dose response regardless of prior COVID infection, suggesting a need for clinically guided vaccine regimens.
Collapse
Affiliation(s)
- Min Zhao
- Department of Medicine, Division of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Amar H Sheth
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Lauren Pischel
- Department of Medicine, Division of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Yale School of Public Health, New Haven, Connecticut, USA
| | - Tassos C Kyriakides
- Department of Veterans Affairs Office of Research and Development, Cooperative Studies Program Coordinating Center, West Haven, Connecticut, USA
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Brinda Emu
- Department of Medicine, Division of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Division of Infectious Diseases, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
| | - Cynthia McNamara
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Qiaosu Shi
- Department of Medicine, Division of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jaden Delgobbo
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- University of Connecticut, Storrs, Connecticut, USA
| | - Jin Xu
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Elizabeth Marhoffer
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aleagia Mercer-Falkoff
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jürgen Holleck
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - David Ardito
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
| | - Richard E Sutton
- Department of Medicine, Division of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Division of Infectious Diseases, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
| | - Shaili Gupta
- Department of Medicine, Division of Infectious Diseases, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Veterans Affairs Healthcare Systems of Connecticut, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Xenos A, Malod-Dognin N, Zambrana C, Pržulj N. Integrated Data Analysis Uncovers New COVID-19 Related Genes and Potential Drug Re-Purposing Candidates. Int J Mol Sci 2023; 24:1431. [PMID: 36674947 PMCID: PMC9863794 DOI: 10.3390/ijms24021431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
The COVID-19 pandemic is an acute and rapidly evolving global health crisis. To better understand this disease's molecular basis and design therapeutic strategies, we built upon the recently proposed concept of an integrated cell, iCell, fusing three omics, tissue-specific human molecular interaction networks. We applied this methodology to construct infected and control iCells using gene expression data from patient samples and three cell lines. We found large differences between patient-based and cell line-based iCells (both infected and control), suggesting that cell lines are ill-suited to studying this disease. We compared patient-based infected and control iCells and uncovered genes whose functioning (wiring patterns in iCells) is altered by the disease. We validated in the literature that 18 out of the top 20 of the most rewired genes are indeed COVID-19-related. Since only three of these genes are targets of approved drugs, we applied another data fusion step to predict drugs for re-purposing. We confirmed with molecular docking that the predicted drugs can bind to their predicted targets. Our most interesting prediction is artenimol, an antimalarial agent targeting ZFP62, one of our newly identified COVID-19-related genes. This drug is a derivative of artemisinin drugs that are already under clinical investigation for their potential role in the treatment of COVID-19. Our results demonstrate further applicability of the iCell framework for integrative comparative studies of human diseases.
Collapse
Affiliation(s)
- Alexandros Xenos
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, Universitat Politecnica de Catalunya (UPC), 08034 Barcelona, Spain
| | - Noël Malod-Dognin
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Carme Zambrana
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, Universitat Politecnica de Catalunya (UPC), 08034 Barcelona, Spain
| | - Nataša Pržulj
- Barcelona Supercomputing Center (BSC), 08034 Barcelona, Spain
- Department of Computer Science, University College London, London WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
19
|
Ghaleh SS, Rahimian K, Mahmanzar M, Mahdavi B, Tokhanbigli S, Sisakht MM, Farhadi A, Bakhtiari MM, Kuehu DL, Deng Y. SARS-CoV-2 Non-structural protein 1(NSP1) mutation virulence and natural selection: Evolutionary trends in the six continents. Virus Res 2023; 323:199016. [PMID: 36473671 PMCID: PMC9721189 DOI: 10.1016/j.virusres.2022.199016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Rapid transmission and reproduction of RNA viruses prepare conducive conditions to have a high rate of mutations in their genetic sequence. The viral mutations make adapt the severe acute respiratory syndrome coronavirus 2 in the host environment and help the evolution of the virus then also caused a high mortality rate by the virus that threatens worldwide health. Mutations and adaptation help the virus to escape confrontations that are done against it. METHODS In the present study, we analyzed 6,510,947 sequences of non-structural protein 1 as one of the conserved regions of the virus to find out frequent mutations and substitute amino acids in comparison with the wild type. NSP1 mutations rate divided into continents were different. RESULTS Based on this continental categorization, E87D in global vision and also in Europe notably increased. The E87D mutation has signed up to January 2022 as the first frequent mutation observed. The remarkable mutations, H110Y and R24C have the second and third frequencies, respectively. CONCLUSION According to the important role of non-structural protein 1 on the host mRNA translation, developing drug design against the protein could be so hopeful to find more effective ways the control and then treatment of the global pandemic coronavirus disease 2019.
Collapse
Affiliation(s)
| | - Karim Rahimian
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics. Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammadamin Mahmanzar
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Tokhanbigli
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran (IAUPS)
| | - Mahsa Mollapour Sisakht
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Amin Farhadi
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Mahsa Mousakhan Bakhtiari
- Pediatric Cell Therapy Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Donna Lee Kuehu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA.
| |
Collapse
|
20
|
Anderson BD, Barnes AN, Umar S, Guo X, Thongthum T, Gray GC. Reverse Zoonotic Transmission (Zooanthroponosis): An Increasing Threat to Animal Health. ZOONOSES: INFECTIONS AFFECTING HUMANS AND ANIMALS 2023:25-87. [DOI: 10.1007/978-3-031-27164-9_59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
21
|
Kocyigit A, Guler EM, Irban A, Kiran B, Atayoglu AT. Assessment of Association Between the Potential Immunomodulatory Activity and Drinking Olive Leaf Tea in the Coronavirus Disease-2019 Pandemic: An Observational Study. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2022; 28:940-947. [PMID: 36112183 DOI: 10.1089/jicm.2022.0554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: During the Coronavirus Disease-2019 (COVID-19) pandemic, in addition to the current measures, the healthy immune system plays an essential role and various natural agents have been recommended to boost innate immunity. The aim of this study was to investigate any association between the potential immunomodulatory activity and drinking olive leaf tea (OLT) in the COVID-19 pandemic. Design: The study was conducted among the workers in a tractor factory where OLT was served in routine. Drinking at least one cup of OLT per day for a minimum of 1 month was the inclusion criteria used in the study. The workers who had a history of vaccination and COVID-19 were excluded from the study, and lymphocyte subsets, interleukin (IL)-2, IFN-γ, COVID-19-specific IgM and IgG levels were analyzed in all the participants to determine the asymptomatic individuals among the participants and compare the immunological parameters. Results: The study was conducted among 336 workers, 183 of them were OLT drinkers and 153 were OLT nondrinkers. The results showed higher values of CD3-/CD16/56 (natural killer [NK]) cells, CD3+/CD16/56 (natural killer T [NKT]) cells, total NK (NK+NKT) cells, and serum IFN-γ, and IL-2 levels in OLT drinkers compared to the nondrinkers. Although all the OLT drinkers and nondrinkers included in the study reported no history of COVID-19, specific COVID-19 IgG levels were found positive in 60% of OLT drinkers and 38% OLT nondrinkers. Conclusions: Peripheral NK and NKT cell values and IL-2 and IFN-γ secretion levels were found higher in the OLT drinking group. There were positive correlations between the OLT drinking frequency and NK cell counts. Moreover, the number of individuals who had "asymptomatic" COVID-19 infection was higher in the OLT drinking group than in the nondrinking cohort. Clinical Trial Registration Number: The trial has been registered in the ClinicalTrials.gov database (CTR NCT05222347).
Collapse
Affiliation(s)
- Abdurrahim Kocyigit
- Department of Biochemistry, Faculty of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Eray Metin Guler
- Department of Clinical Biochemistry and Hamidiye Medicine Faculty, Health Sciences University, Istanbul, Turkey
| | - Arzu Irban
- Department of Anestesia and Reanimation, Hamidiye Medicine Faculty, Health Sciences University, Istanbul, Turkey
| | - Bayram Kiran
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Kastamonu University, Kastamonu, Turkey
| | - Ali Timucin Atayoglu
- Department of Family Medicine, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
22
|
Sarangi MK, Padhi S, Rath G, Nanda SS, Yi DK. Success of nano-vaccines against COVID-19: a transformation in nanomedicine. Expert Rev Vaccines 2022; 21:1739-1761. [PMID: 36384360 DOI: 10.1080/14760584.2022.2148659] [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: 11/18/2022]
Abstract
INTRODUCTION The vaccines being used against COVID-19 are composed of either non-viral or viral nanoparticles (NPs). Nanotechnology-based vaccine technology was studied for its potentially transformative advancement of medicine. AREAS COVERED NPs protect the encapsulated mRNA in vaccines, thereby enhancing the stability of the ribonucleic acids and facilitating their intact delivery to their specific targets. Compared to liposomes, lipid nanoparticles (LNPs) are unique and, through their rigid morphology and better cellular penetrability, render enhanced cargo stability. To explore nanotechnology-mediated vaccine delivery and its potential in future pandemics, we assessed articles from various databases, such as PubMed, Embase, and Scopus, including editorial/research notes, expert opinions, and collections of data from several clinical research trials. In the current review, we focus on the nanoparticulate approach of the different SARS-CoV-2 vaccines and explore their success against the pandemic. EXPERT OPINION The mRNA-based vaccines, with their tremendous efficacy of ~95% (under phase III-IV clinical trials) and distinct nanocarriers (LNPs), represent a new medical front alongside DNA and siRNA-based vaccines.
Collapse
Affiliation(s)
- Manoj Kumar Sarangi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Sasmita Padhi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Gautam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | | | - Dong Kee Yi
- Department of Chemistry, Myongji University, Yongin, South Korea
| |
Collapse
|
23
|
Mohseni Afshar Z, Barary M, Hosseinzadeh R, Karim B, Ebrahimpour S, Nazary K, Sio TT, Sullman MJM, Carson-Chahhoud K, Moudi E, Babazadeh A. COVID-19 vaccination challenges: A mini-review. Hum Vaccin Immunother 2022; 18:2066425. [PMID: 35512088 PMCID: PMC9302531 DOI: 10.1080/21645515.2022.2066425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/11/2022] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
The emergence of SARS-CoV-2 has led to the infection of many people across the globe, over six million deaths, and has placed an unprecedented burden on public health worldwide. The pandemic has led to the high-speed development and production of vaccines against the COVID-19, as vaccines can end the pandemic. At the beginning of the program, vaccinations were initially targeted only at high-risk groups, such as the elderly, those with comorbidities, or healthcare workers. Although most of the mentioned populations have received the two recommended doses, limited resources have left many authorities with an effective vaccine undersupply. Therefore, policies have been implemented to manage the available doses of the vaccines more efficiently. As there is no universally agreed consensus on this topic, we discuss the different recommendations and guidelines regarding the time interval between the two vaccine doses and explain the different scenarios for applying the two doses.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kosar Nazary
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Terence T. Sio
- Department of Radiation Oncology, Mayo Clinic, Phoenix, Arizona, USA
| | - Mark J. M. Sullman
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | | | - Emaduddin Moudi
- Clinical Research Development Center, Shahid Beheshti Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
24
|
Dhama K, Dhawan M, Tiwari R, Emran TB, Mitra S, Rabaan AA, Alhumaid S, Alawi ZA, Al Mutair A. COVID-19 intranasal vaccines: current progress, advantages, prospects, and challenges. Hum Vaccin Immunother 2022; 18:2045853. [PMID: 35258416 PMCID: PMC8935456 DOI: 10.1080/21645515.2022.2045853] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Multiple vaccines have recently been developed, and almost all the countries are presently vaccinating their population to tackle the COVID-19 pandemic. Most of the COVID-19 vaccines in use are administered via intramuscular (IM) injection, eliciting protective humor and cellular immunity. COVID-19 intranasal (IN) vaccines are also being developed that have shown promising ability to induce a significant amount of antibody-mediated immune response and a robust cell-mediated immunity as well as hold the added ability to stimulate protective mucosal immunity along with the additional advantage of the ease of administration as compared to IM injected vaccines. By inducing secretory IgA antibody responses specifically in the nasal compartment, the intranasal SARS-CoV-2 vaccine can prevent virus infection, replication, shedding, and disease development, as well as possibly limits virus transmission. This article highlights the current progress, advantages, prospects, and challenges in developing intranasal COVID-19 vaccines for countering the ongoing pandemic.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- The Trafford Group of Colleges, Manchester, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Talha Bin Emran
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa, Saudi Arabia
| | - Zainab Al Alawi
- Division of Allergy and Immunology, College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, Australia
| |
Collapse
|
25
|
A Quantum Vaccinomics Approach for the Design and Production of MSP4 Chimeric Antigen for the Control of Anaplasma phagocytophilum Infections. Vaccines (Basel) 2022; 10:vaccines10121995. [PMID: 36560405 PMCID: PMC9784196 DOI: 10.3390/vaccines10121995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Anaplasma phagocytophilum Major surface protein 4 (MSP4) plays a role during infection and multiplication in host neutrophils and tick vector cells. Recently, vaccination trials with the A. phagocytophilum antigen MSP4 in sheep showed only partial protection against pathogen infection. However, in rabbits immunized with MSP4, this recombinant antigen was protective. Differences between rabbit and sheep antibody responses are probably associated with the recognition of non-protective epitopes by IgG of immunized lambs. To address this question, we applied quantum vaccinomics to identify and characterize MSP4 protective epitopes by a microarray epitope mapping using sera from vaccinated rabbits and sheep. The identified candidate protective epitopes or immunological quantum were used for the design and production of a chimeric protective antigen. Inhibition assays of A. phagocytophilum infection in human HL60 and Ixodes scapularis tick ISE6 cells evidenced protection by IgG from sheep and rabbits immunized with the chimeric antigen. These results supported that the design of new chimeric candidate protective antigens using quantum vaccinomics to improve the protective capacity of antigens in multiple hosts.
Collapse
|
26
|
Garnett L, Tse C, Funk D, Dust K, Tran KN, Hedley A, Poliquin G, Bullard J, Strong JE. Differential Infectivity of Original and Delta Variants of SARS-CoV-2 in Children Compared to Adults. Microbiol Spectr 2022; 10:e0039522. [PMID: 35972128 PMCID: PMC9602606 DOI: 10.1128/spectrum.00395-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Although children of all ages are susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, they have not been implicated as major drivers of transmission thus far. However, it is still unknown if this finding holds true with new variants of concern (VOC), such as Delta (B.1.617.2). This study aimed to examine differences in both viral RNA (as measured by cycle threshold [CT]) and viable-virus levels from children infected with Delta and those infected with original variants (OV). Furthermore, we aimed to compare the pediatric population infection trends to those in adults. We obtained 690 SARS-CoV-2 RT-PCR positive nasopharyngeal swabs from across Manitoba, Canada, which were further screened for mutations characteristic of VOC. Aliquots of sample were then provided for TCID50 (50% tissue culture infective dose) assays to determine infectious titers. Using a variety of statistical analyses we compared CT and infectivity of VOC in different age demographics. Comparing 122 Delta- to 175 OV-positive nasopharyngeal swab samples from children, we found that those infected with Delta are 2.7 times more likely to produce viable SARS-CoV-2 with higher titers (in TCID50 per milliliter), regardless of viral RNA levels. Moreover, comparing the pediatric samples to 130 OV- and 263 Delta-positive samples from adults, we found only that the Delta pediatric culture-positive samples had titers (TCID50 per milliliter) similar to those of culture-positive adult samples. IMPORTANCE These important findings show that children may play a larger role in viral transmission of Delta than for previously circulating SARS-CoV-2 variants. Additionally, they may suggest a mechanism for why Delta has evolved to be the predominant circulating variant.
Collapse
Affiliation(s)
- Lauren Garnett
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Carmen Tse
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Duane Funk
- Departments of Anaesthesiology and Medicine, Section of Critical Care, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kerry Dust
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Manitoba, Canada
| | - Kaylie N. Tran
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Adam Hedley
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Manitoba, Canada
| | - Guillaume Poliquin
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jared Bullard
- Cadham Provincial Laboratory, Manitoba Health, Winnipeg, Manitoba, Canada
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James E. Strong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
27
|
Analysis of the mutation dynamics of SARS-CoV-2 genome in the samples from Georgia State of the United States. Gene 2022; 841:146774. [PMID: 35905853 PMCID: PMC9323210 DOI: 10.1016/j.gene.2022.146774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/12/2022] [Accepted: 07/24/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The COVID-19 is caused by a novel coronavirus SARS-CoV-2, which started from China. It spread rapidly throughout the world and was later declared a pandemic by the WHO. Over the course of time, SARS-CoV-2 has mutated for survival advantages, and this led to multiple variants. Multiple studies on mutations identification in SARS-CoV2 have been published covering extensive sample areas. The purpose of this study was to limit the sample area to the Georgia state in the U.S. and to analyze the genome sequences for mutation profiling across the genome and origin of variants. METHODS The genome sequences (n = 3,970) were obtained from the NCBI database as of June 12, 2021, with the filter of being complete sequenced genomes, homo-sapiens host, and only from Georgia State of the U.S. NextClade, an online tool was used for the analysis of the sequences using Wuhan-Hu-1/2019 as a reference genome. The algorithm was sequence alignment, translation, mutation calling, phylogenetic placement, clade assignment, and quality control (QC). Thirty-six samples with bad QC were removed from the mutational analysis. RESULTS A total 117,743 mutations in the nucleotides were identified (averaging 31.5 mutations per sample). The mutations A23403G, C3037T, C241T, and C14408T were detected in 98% of the samples. Also, a total of 75,517 mutations in the amino acid were identified (averaging 20.2 mutations per sample). The mutations D614G and P314L were identified in >97% samples whereas R203K, G204R, P681H, and N501Y were detected in >50% samples. Analysis also revealed 16 different clades with 20I (49.6%). Clades 20G (24.2%) and 20A (5.5%) being the most abundant, showed that SARS-CoV-2 in the Georgia State originated mainly from Southeast England, other parts of the U.S., and several countries in Western Europe. CONCLUSION Looking at the three most common variants in Georgia State of the U.S., we could determine the primary locations of transmission or origin for the virus, and our analyses indicates that majority of the cases originated from Southeast England (Clade 20I), the U.S. itself (Clade 20G), and from Western Europe (Clade 20C).
Collapse
|
28
|
Noor R. How do the severe acute respiratory coronavirus 2 (SARS-CoV-2) and its variants escape the host protective immunity and mediate pathogenesis? BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:255. [PMID: 36254244 PMCID: PMC9556142 DOI: 10.1186/s42269-022-00945-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/06/2022] [Indexed: 05/10/2023]
Abstract
Background To protect the global population from the ongoing COVID-19 pandemic caused by the severe acute respiratory β-coronavirus 2 (SARS-CoV-2), a number of vaccines are currently being used in three dosages (i.e., along with the booster dose) to induce the immunity required to combat the SARS-CoV-2 and its variants. So far, several antivirals and the commercial vaccines have been found to evoke the required humoral and cellular immunity within a huge population around world. However, an important aspect to consider is the avoidance mechanism of the host protective immunity by SARS-CoV-2 variants. Main body of the abstract Indeed, such an immune escape strategy has been noticed previously in case of SARS-CoV-1 and the Middle East Respiratory Syndrome coronavirus (MERS-CoV). Regarding the SARS-CoV-2 variants, the most important aspect on vaccine development is to determine whether the vaccine is actually capable to elicit the immune response or not, especially the viral spike (S) protein. Short conclusion Present review thus focused on such elicitation of immunity as well as pondered to the avoidance of host immunity by the SARS-CoV-2 Wuhan strain and its variants.
Collapse
Affiliation(s)
- Rashed Noor
- Department of Life Sciences (DLS), School of Environment and Life Sciences (SELS), Independent University, Bangladesh (IUB), Plot 16, Block B, Aftabuddin Ahmed Road, Bashundhara, Dhaka 1229 Bangladesh
| |
Collapse
|
29
|
Daher-Nashif S, Al-Anany R, Ali M, Erradi K, Farag E, Abdallah AM, Emara MM. COVID-19 exit strategy during vaccine implementation: a balance between social distancing and herd immunity. Arch Virol 2022; 167:1773-1783. [PMID: 35723757 PMCID: PMC9208258 DOI: 10.1007/s00705-022-05495-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/05/2022] [Indexed: 11/09/2022]
Abstract
Currently, health authorities around the world are struggling to limit the spread of COVID-19. Since the beginning of the pandemic, social distancing has been the most important strategy used by most countries to control disease spread by flattening and elongating the epidemic curve. Another strategy, herd immunity, was also applied by some countries through relaxed control measures that allow the free spread of natural infection to build up solid immunity within the population. In 2021, COVID-19 vaccination was introduced with tremendous effort as a promising strategy for limiting the spread of disease. Therefore, in this review, we present the current knowledge about social distancing, herd immunity strategies, and aspects of their implementation to control the COVID-19 pandemic in the presence of the newly developed vaccines. Finally, we suggest a short-term option for controlling the pandemic during vaccine application.
Collapse
Affiliation(s)
- Suhad Daher-Nashif
- Population Medicine Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rania Al-Anany
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Public Health Department, Health Protection and Communicable Diseases, Ministry of Public Health, Doha, Qatar
| | - Menatalla Ali
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Khadija Erradi
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Elmoubasher Farag
- Public Health Department, Health Protection and Communicable Diseases, Ministry of Public Health, Doha, Qatar
| | - Abdallah M Abdallah
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M Emara
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
30
|
Muñoz-Jurado A, Escribano BM, Agüera E, Caballero-Villarraso J, Galván A, Túnez I. SARS-CoV-2 infection in multiple sclerosis patients: interaction with treatments, adjuvant therapies, and vaccines against COVID-19. J Neurol 2022; 269:4581-4603. [PMID: 35788744 PMCID: PMC9253265 DOI: 10.1007/s00415-022-11237-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022]
Abstract
The SARS-CoV-2 pandemic has raised particular concern for people with Multiple Sclerosis, as these people are believed to be at increased risk of infection, especially those being treated with disease-modifying therapies. Therefore, the objective of this review was to describe how COVID-19 affects people who suffer from Multiple Sclerosis, evaluating the risk they have of suffering an infection by this virus, according to the therapy to which they are subjected as well as the immune response of these patients both to infection and vaccines and the neurological consequences that the virus can have in the long term. The results regarding the increased risk of infection due to treatment are contradictory. B-cell depletion therapies may cause patients to have a lower probability of generating a detectable neutralizing antibody titer. However, more studies are needed to help understand how this virus works, paying special attention to long COVID and the neurological symptoms that it causes.
Collapse
Affiliation(s)
- Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Campus of Rabanales, 14071 Cordoba, Spain
| | - Begoña M. Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Campus of Rabanales, 14071 Cordoba, Spain
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
| | - Eduardo Agüera
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Neurology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Javier Caballero-Villarraso
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
- Clinical Analysis Service, Reina Sofía University Hospital, Cordoba, Spain
| | - Alberto Galván
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
| | - Isaac Túnez
- Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBC), Cordoba, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Av. Menendez Pidal, 14004 Cordoba, Spain
- Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Madrid, Spain
| |
Collapse
|
31
|
Salami Ghaleh S, Rahimian K, Mahmanzar M, Mahdavi B, Tokhanbigli S, Mollapour Sisakht M, Farhadi A, Mousakhan Bakhtiari M, Lee Kuehu D, Deng Y. SARS-CoV-2 Non-Structural Protein 1(NSP1) Mutation Virulence and Natural Selection: Evolutionary Trends in the Six Continents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.07.22.501212. [PMID: 35923310 PMCID: PMC9347281 DOI: 10.1101/2022.07.22.501212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an unsegmented positivesense single-stranded RNA virus that belongs to the β-coronavirus . This virus was the cause of a novel severe acute respiratory syndrome in 2019 (COVID-19) that emerged in Wuhan, China at the early stage of the pandemic and rapidly spread around the world. Rapid transmission and reproduction of SARS-CoV-2 threaten worldwide health with a high mortality rate from the virus. According to the significant role of non-structural protein 1 (NSP1) in inhibiting host mRNA translation, this study focuses on the link between amino acid sequences of NSP1 and alterations of them spreading around the world. The SARS-CoV-2 NSP1 protein sequences were analyzed and FASTA files were processed by Python language programming libraries. Reference sequences compared with each NSP1 sample to identify every mutation and categorize them were based on continents and frequencies. NSP1 mutations rate divided into continents were different. Based on continental studies, E87D in global vision and also in Europe notably increased. The E87D mutation has significantly risen especially in the last months of the study as the first frequent mutation observed. The remarkable mutations, H110Y and R24C, have the second and third frequencies, respectively. Based on this mutational information, despite NSP1 being a conserved sequence occurrence, these mutations change the rate of flexibility and stability of the NSP1 protein, which can eventually affect inhibiting the host translation. IMPORTANCE In this study, we analyzed 6,510,947 sequences of non-structural protein 1 as a conserved region of SARS-CoV-2. According to the obtained results, 93.4819% of samples had no mutant regions on their amino acid sequences. Heat map data of mutational samples demonstrated high percentages of mutations that occurred in the region of 72 to 126 amino acids indicating a hot spot region of the protein. Increased rates of E87D, H110Y, and R24C mutations in the timeline of our study were reported as significant compared to available mutant samples. Analyzing the details of replacing amino acids in the most frequent E87D mutation reveals the role of this alteration in increasing molecule flexibility and destabilizing the structure of the protein.
Collapse
|
32
|
Miranda RR, Ferreira NN, Souza EED, Lins PMP, Ferreira LM, Krüger A, Cardoso VMD, Durigon EL, Wrenger C, Zucolotto V. Modulating Fingolimod (FTY720) Anti-SARS-CoV-2 Activity Using a PLGA-Based Drug Delivery System. ACS APPLIED BIO MATERIALS 2022; 5:3371-3383. [PMID: 35732506 PMCID: PMC9236206 DOI: 10.1021/acsabm.2c00349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/12/2022] [Indexed: 11/30/2022]
Abstract
COVID-19 has resulted in more than 490 million people being infected worldwide, with over 6 million deaths by April 05th, 2022. Even though the development of safe vaccine options is an important step to reduce viral transmission and disease progression, COVID-19 cases will continue to occur, and for those cases, efficient treatment remains to be developed. Here, a drug repurposing strategy using nanotechnology is explored to develop a therapy for COVID-19 treatment. Nanoparticles (NPs) based on PLGA for fingolimod (FTY720) encapsulation show a size of ∼150 nm and high drug entrapment (∼90%). The NP (NP@FTY720) can control FTY720 release in a pH-dependent manner. Cytotoxicity assays using different cell lines show that NP@FTY720 displays less toxicity than the free drug. Flow cytometry and confocal microscopy reveal that NPs are actively internalized mostly through caveolin-mediated endocytosis and macropinocytosis pathways and co-localized with lysosomes. Finally, NP@FTY720 not only exhibits anti-SARS-CoV-2 activity at non-cytotoxic concentrations, but its biological potential for viral infection inhibition is nearly 70 times higher than that of free drug treatment. Based on these findings, the combination of drug repurposing and nanotechnology as NP@FTY720 is presented for the first time and represents a promising frontline in the fight against COVID-19.
Collapse
Affiliation(s)
- Renata Rank Miranda
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| | - Natália Noronha Ferreira
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| | - Edmarcia Elisa de Souza
- Unit for Drug Discovery, Department of Parasitology,
Institute of Biomedical Sciences, University of Sao Paulo, Av.
Prof. Lineu Prestes 1374, 05508-000 Sao Paulo, Sao Paulo, Brazil
| | - Paula Maria Pincela Lins
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| | - Leonardo Miziara
Barboza Ferreira
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| | - Arne Krüger
- Unit for Drug Discovery, Department of Parasitology,
Institute of Biomedical Sciences, University of Sao Paulo, Av.
Prof. Lineu Prestes 1374, 05508-000 Sao Paulo, Sao Paulo, Brazil
| | - Valéria Maria de
Oliveira Cardoso
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| | - Edison Luiz Durigon
- Unit for Drug Discovery, Department of Parasitology,
Institute of Biomedical Sciences, University of Sao Paulo, Av.
Prof. Lineu Prestes 1374, 05508-000 Sao Paulo, Sao Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology,
Institute of Biomedical Sciences, University of Sao Paulo, Av.
Prof. Lineu Prestes 1374, 05508-000 Sao Paulo, Sao Paulo, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, Physics
Institute of São Carlos, São Paulo University,
Avenida Trabalhador São Carlense, 400, 13566-590 São Carlos, São
Paulo, Brazil
| |
Collapse
|
33
|
Hirsh J, Htay T, Bhalla S, Nguyen V, Cervantes J. Breakthrough SARS-CoV-2 infections after COVID-19 immunization. J Investig Med 2022; 70:1429-1432. [PMID: 35768140 DOI: 10.1136/jim-2021-002131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 12/15/2022]
Abstract
As no vaccines are 100% effective at preventing illness, COVID-19 vaccine breakthrough cases are expected. We here aim to review the most recent literature on COVID-19 vaccine breakthrough infections. SARS-CoV-2 breakthrough infections are, in general, rare. Age may still be a factor in SARS-CoV-2 infections in immunized individuals.
Collapse
Affiliation(s)
- Joshua Hirsh
- Medical Education, Texas Tech University Health Sciences Center El Paso Paul L Foster School of Medicine, El Paso, Texas, USA
| | - Thwe Htay
- Medical Education, Texas Tech University Health Sciences Center El Paso Paul L Foster School of Medicine, El Paso, Texas, USA
| | - Shubhang Bhalla
- Medical Education, Texas Tech University Health Sciences Center El Paso Paul L Foster School of Medicine, El Paso, Texas, USA
| | - Victoria Nguyen
- Medical Education, Texas Tech University Health Sciences Center El Paso Paul L Foster School of Medicine, El Paso, Texas, USA
| | - Jorge Cervantes
- Medical Education, Texas Tech University Health Sciences Center El Paso Paul L Foster School of Medicine, El Paso, Texas, USA
| |
Collapse
|
34
|
Das S, Kar SS, Samanta S, Banerjee J, Giri B, Dash SK. Immunogenic and reactogenic efficacy of Covaxin and Covishield: a comparative review. Immunol Res 2022; 70:289-315. [PMID: 35192185 PMCID: PMC8861611 DOI: 10.1007/s12026-022-09265-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 is an RNA virus that was identified for the first time in December 2019 in Wuhan, China. The World Health Organization (WHO) labeled the novel coronavirus (COVID-19) outbreak a worldwide pandemic on March 11, 2020, due to its widespread infectivity pattern. Because of the catastrophic COVID-19 outbreak, the development of safe and efficient vaccinations has become a key priority in every health sector throughout the globe. On the 13th of January 2021, the vaccination campaign against SARS-CoV-2 was launched in India and started the administration of two types of vaccines known as Covaxin and Covishield. Covishield is an adenovirus vector-based vaccine, and Covaxin was developed by a traditional method of vaccine formulation, which is composed of adjuvanted inactivated viral particles. Each vaccine's utility or efficiency is determined by its formulation, adjuvants, and mode of action. The efficacy of the vaccination depends on numeral properties like generation antibodies, memory cells, and cell-mediated immunity. According to the third-phase experiment, Covishield showed effectiveness of nearly 90%, whereas Covaxin has an effectiveness of about 80%. Both vaccination formulations in India have so far demonstrated satisfactory efficacy against numerous mutant variants of SARS-CoV-2. The efficacy of Covishield may be diminished if the structure of spike (S) protein changes dramatically in the future. In this situation, Covaxin might be still effective for such variants owing to its ability to produce multiple antibodies against various epitopes. This study reviews the comparative immunogenic and therapeutic efficacy of Covaxin and Covishield and also discussed the probable vaccination challenges in upcoming days.
Collapse
Affiliation(s)
- Swarnali Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Suvrendu Sankar Kar
- Department of Medicine, R.G.Kar Medical College, Kolkata, 700004, West Bengal, India
| | - Sovan Samanta
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India.
| |
Collapse
|
35
|
Zhao M, Slotkin R, Sheth AH, Pischel L, Kyriakides TC, Emu B, McNamara C, Shi Q, Delgobbo J, Xu J, Marhoffer E, Mercer-Falkoff A, Holleck J, Ardito D, Sutton RE, Gupta S. Clinical Variables Correlate with Serum Neutralizing Antibody Titers after COVID-19 mRNA Vaccination in an Adult, US-based Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.04.03.22273355. [PMID: 35411347 PMCID: PMC8996621 DOI: 10.1101/2022.04.03.22273355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background We studied whether comorbid conditions impact strength and duration of immune responses after SARS-CoV-2 mRNA vaccination in a US-based, adult population. Methods Sera (pre-and-post-BNT162b2 vaccination) were tested serially up to 12 months after two doses of vaccine for SARS-CoV-2-anti-Spike neutralizing capacity by pseudotyping assay in 124 individuals; neutralizing titers were correlated to clinical variables with multivariate regression. Post-booster (third dose) effect was measured at 1 and 3 months in 72 and 88 subjects respectively. Results After completion of primary vaccine series, neutralizing antibody IC50 values were high at one month (14-fold increase from pre-vaccination), declined at six months (3.3-fold increase), and increased at one month post-booster (41.5-fold increase). Three months post-booster, IC50 decreased in COVID-naïve individuals (18-fold increase) and increased in prior COVID-19+ individuals (132-fold increase). Age >65 years (β=-0.94, p=0.001) and malignancy (β=-0.88, p=0.002) reduced strength of response at 1 month. Both strength and durability of response at 6 months, respectively, were negatively impacted by end-stage renal disease [(β=-1.10, p=0.004); (β=-0.66, p=0.014)], diabetes mellitus [(β=-0.57, p=0.032); (β=-0.44, p=0.028)], and systemic steroid use [(β=-0.066, p=0.032); (β=-0.55, p=0.037)]. Post-booster IC50 was robust against WA-1 and B.1.617.2, but the immune response decreased with malignancy (β =-0.68, p=0.03) and increased with prior COVID-19 (p-value < 0.0001). Conclusion Multiple clinical factors impact the strength and duration of neutralization response post-primary series vaccination, but not the post-booster dose strength. Prior COVID-19 infection enhances the booster-dose response except in individuals with malignancy, suggesting a need for clinically guiding vaccine dosing regimens. Summary Multiple clinical factors impact the strength and duration of neutralization response post-primary series vaccination. All subjects, irrespective of prior COVID infection, benefited from a third dose. Malignancy decreased response following third dose, suggesting the importance of clinically guided vaccine regimens.
Collapse
|
36
|
Molteni E, Sudre CH, Canas LDS, Bhopal SS, Hughes RC, Chen L, Deng J, Murray B, Kerfoot E, Antonelli M, Graham M, Kläser K, May A, Hu C, Pujol JC, Wolf J, Hammers A, Spector TD, Ourselin S, Modat M, Steves CJ, Absoud M, Duncan EL. Illness Characteristics of COVID-19 in Children Infected with the SARS-CoV-2 Delta Variant. CHILDREN (BASEL, SWITZERLAND) 2022; 9:652. [PMID: 35626830 PMCID: PMC9140086 DOI: 10.3390/children9050652] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND The Delta (B.1.617.2) SARS-CoV-2 variant was the predominant UK circulating strain between May and November 2021. We investigated whether COVID-19 from Delta infection differed from infection with previous variants in children. METHODS Through the prospective COVID Symptom Study, 109,626 UK school-aged children were proxy-reported between 28 December 2020 and 8 July 2021. We selected all symptomatic children who tested positive for SARS-CoV-2 and were proxy-reported at least weekly, within two timeframes: 28 December 2020 to 6 May 2021 (Alpha (B.1.1.7), the main UK circulating variant) and 26 May to 8 July 2021 (Delta, the main UK circulating variant), with all children unvaccinated (as per national policy at the time). We assessed illness profiles (symptom prevalence, duration, and burden), hospital presentation, and presence of long (≥28 day) illness, and calculated odds ratios for symptoms presenting within the first 28 days of illness. RESULTS 694 (276 younger (5-11 years), 418 older (12-17 years)) symptomatic children tested positive for SARS-CoV-2 with Alpha infection and 706 (227 younger and 479 older) children with Delta infection. Median illness duration was short with either variant (overall cohort: 5 days (IQR 2-9.75) with Alpha, 5 days (IQR 2-9) with Delta). The seven most prevalent symptoms were common to both variants. Symptom burden over the first 28 days was slightly greater with Delta compared with Alpha infection (in younger children, 3 (IQR 2-5) symptoms with Alpha, 4 (IQR 2-7) with Delta; in older children, 5 (IQR 3-8) symptoms with Alpha, 6 (IQR 3-9) with Delta infection ). The odds of presenting several symptoms were higher with Delta than Alpha infection, including headache and fever. Few children presented to hospital, and long illness duration was uncommon, with either variant. CONCLUSIONS COVID-19 in UK school-aged children due to SARS-CoV-2 Delta strain B.1.617.2 resembles illness due to the Alpha variant B.1.1.7., with short duration and similar symptom burden.
Collapse
Affiliation(s)
- Erika Molteni
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Carole H. Sudre
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
- MRC Unit for Lifelong Health and Ageing, Department of Population Health Sciences, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Liane Dos Santos Canas
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Sunil S. Bhopal
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Robert C. Hughes
- Department of Population Health, Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Liyuan Chen
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Jie Deng
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Benjamin Murray
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Eric Kerfoot
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Michela Antonelli
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Mark Graham
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Kerstin Kläser
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Anna May
- ZOE Limited London, London SE1 7RW, UK; (A.M.); (C.H.); (J.C.P.); (J.W.)
| | - Christina Hu
- ZOE Limited London, London SE1 7RW, UK; (A.M.); (C.H.); (J.C.P.); (J.W.)
| | | | - Jonathan Wolf
- ZOE Limited London, London SE1 7RW, UK; (A.M.); (C.H.); (J.C.P.); (J.W.)
| | - Alexander Hammers
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
- King’s College London & Guy’s and St Thomas’ PET Centre, London WC2R 2LS, UK
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London WC2R 2LS, UK; (T.D.S.); (C.J.S.)
| | - Sebastien Ourselin
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London WC2R 2LS, UK; (E.M.); (C.H.S.); (L.D.S.C.); (L.C.); (J.D.); (B.M.); (E.K.); (M.A.); (M.G.); (K.K.); (A.H.); (S.O.); (M.M.)
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, King’s College London, London WC2R 2LS, UK; (T.D.S.); (C.J.S.)
- Department of Aging and Health, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK
| | - Michael Absoud
- Children’s Neurosciences, Evelina London Children’s Hospital, St Thomas’ Hospital, King’s Health Partners, Academic Health Science Centre, London SE1 7EH, UK
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, School of Life Course Sciences, King’s College London, London WC2R 2LS, UK
| | - Emma L. Duncan
- Department of Twin Research and Genetic Epidemiology, King’s College London, London WC2R 2LS, UK; (T.D.S.); (C.J.S.)
- Department of Endocrinology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK
| |
Collapse
|
37
|
Molteni E, Sudre CH, Canas LDS, Bhopal SS, Hughes RC, Chen L, Deng J, Murray B, Kerfoot E, Antonelli M, Graham M, Kläser K, May A, Hu C, Pujol JC, Wolf J, Hammers A, Spector TD, Ourselin S, Modat M, Steves CJ, Absoud M, Duncan EL. Illness Characteristics of COVID-19 in Children Infected with the SARS-CoV-2 Delta Variant. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9050652. [PMID: 35626830 DOI: 10.1101/2021.10.06.21264467] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 05/22/2023]
Abstract
BACKGROUND The Delta (B.1.617.2) SARS-CoV-2 variant was the predominant UK circulating strain between May and November 2021. We investigated whether COVID-19 from Delta infection differed from infection with previous variants in children. METHODS Through the prospective COVID Symptom Study, 109,626 UK school-aged children were proxy-reported between 28 December 2020 and 8 July 2021. We selected all symptomatic children who tested positive for SARS-CoV-2 and were proxy-reported at least weekly, within two timeframes: 28 December 2020 to 6 May 2021 (Alpha (B.1.1.7), the main UK circulating variant) and 26 May to 8 July 2021 (Delta, the main UK circulating variant), with all children unvaccinated (as per national policy at the time). We assessed illness profiles (symptom prevalence, duration, and burden), hospital presentation, and presence of long (≥28 day) illness, and calculated odds ratios for symptoms presenting within the first 28 days of illness. RESULTS 694 (276 younger (5-11 years), 418 older (12-17 years)) symptomatic children tested positive for SARS-CoV-2 with Alpha infection and 706 (227 younger and 479 older) children with Delta infection. Median illness duration was short with either variant (overall cohort: 5 days (IQR 2-9.75) with Alpha, 5 days (IQR 2-9) with Delta). The seven most prevalent symptoms were common to both variants. Symptom burden over the first 28 days was slightly greater with Delta compared with Alpha infection (in younger children, 3 (IQR 2-5) symptoms with Alpha, 4 (IQR 2-7) with Delta; in older children, 5 (IQR 3-8) symptoms with Alpha, 6 (IQR 3-9) with Delta infection ). The odds of presenting several symptoms were higher with Delta than Alpha infection, including headache and fever. Few children presented to hospital, and long illness duration was uncommon, with either variant. CONCLUSIONS COVID-19 in UK school-aged children due to SARS-CoV-2 Delta strain B.1.617.2 resembles illness due to the Alpha variant B.1.1.7., with short duration and similar symptom burden.
Collapse
Affiliation(s)
- Erika Molteni
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Carole H Sudre
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
- MRC Unit for Lifelong Health and Ageing, Department of Population Health Sciences, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Liane Dos Santos Canas
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Sunil S Bhopal
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Robert C Hughes
- Department of Population Health, Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Liyuan Chen
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Jie Deng
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Benjamin Murray
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Eric Kerfoot
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Michela Antonelli
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Mark Graham
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Kerstin Kläser
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Anna May
- ZOE Limited London, London SE1 7RW, UK
| | | | | | | | - Alexander Hammers
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
- King's College London & Guy's and St Thomas' PET Centre, London WC2R 2LS, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Sebastien Ourselin
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
- Department of Aging and Health, Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, UK
| | - Michael Absoud
- Children's Neurosciences, Evelina London Children's Hospital, St Thomas' Hospital, King's Health Partners, Academic Health Science Centre, London SE1 7EH, UK
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London WC2R 2LS, UK
| | - Emma L Duncan
- Department of Twin Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
- Department of Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, UK
| |
Collapse
|
38
|
Di Pietro M, Dono F, Consoli S, Evangelista G, Pozzilli V, Calisi D, Barbone F, Bonanni L, Onofrj M, De Angelis MV, Sensi SL. Cerebral venous thrombosis without thrombocytopenia after a single dose of COVID-19 (Ad26.COV2.S) vaccine injection: a case report. Neurol Sci 2022; 43:2951-2956. [PMID: 35217969 PMCID: PMC8880295 DOI: 10.1007/s10072-022-05965-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/20/2022] [Indexed: 11/28/2022]
Abstract
Background
The coronavirus pandemic became the hard challenge for the modern global health system. To date, vaccination is the best strategy against Sars-Cov-2-related illness. About 3 billions of people received at least one of the approved vaccines. The related adverse events were reported during the various experimental phases, but newer and less common side effects are emerging post-marketing. Vaccine-induced thrombocytopenia with thrombosis (VITT) is one of these insidious adverse reactions and it is considered responsible of venous thrombosis, in both the splanchnic and the cerebral circulation. Although its mechanism has been presumably established, resembling that observed in heparin-induced thrombocytopenia, some venous thromboses seem not to recognize this etiology and their pathogenesis remains unknown. Here we described a case of cerebral venous thrombosis after administration of the Ad26.COV2.S, presenting without thrombocytopenia, paving the way for possible novel causes of this vaccine-induced pathological condition. Case presentation A 45-year-old woman came to our observation for bilateral periorbital headache associated with retro-orbital pain started 8 days after administration of COVID vaccine Jannsen. Ophthalmologic exam showing a bilateral papilledema raised the suspicion of intracranial hypertension. Cerebral magnetic resonance imaging revealed signal alteration with T1-positive contrast enhancement in the right temporal and insular lobes suggestive of cerebral venous thrombosis. The absence of thrombocytopenia and platelet factor 4 (PF-4) antibodies led the clinicians to rule out VITT. The patient was treated successfully with warfarin. Conclusion Venous thrombosis occurring after COVID-19 vaccination represents an adverse event of special interest. Patients with thrombosis and thrombocytopenia appear to be affected by a general thrombophilic state, sustained by an autoimmune mechanism, and show a higher mortality. Thrombosis without thrombocytopenia’s pathogenesis has not yet been clarified, but laboratory data and good response to vitamin K antagonists help clinicians in the differential diagnosis with VITT. Future research will allow us to discover other possible mechanisms and maybe identify a subgroup of patients with a higher risk of developing this medical complication.
Collapse
Affiliation(s)
- Martina Di Pietro
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Fedele Dono
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology - CAST-, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Stefano Consoli
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Giacomo Evangelista
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Valeria Pozzilli
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Dario Calisi
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Filomena Barbone
- Department of Neurology, "SS Annunziata" Hospital, Chieti, Italy
| | - Laura Bonanni
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Stefano L Sensi
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology - CAST-, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
39
|
Martínez-González B, Soria ME, Vázquez-Sirvent L, Ferrer-Orta C, Lobo-Vega R, Mínguez P, de la Fuente L, Llorens C, Soriano B, Ramos R, Cortón M, López-Rodríguez R, García-Crespo C, Gallego I, de Ávila AI, Gómez J, Enjuanes L, Salar-Vidal L, Esteban J, Fernandez-Roblas R, Gadea I, Ayuso C, Ruíz-Hornillos J, Verdaguer N, Domingo E, Perales C. SARS-CoV-2 Point Mutation and Deletion Spectra and Their Association with Different Disease Outcomes. Microbiol Spectr 2022; 10:e0022122. [PMID: 35348367 PMCID: PMC9045161 DOI: 10.1128/spectrum.00221-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
Mutant spectra of RNA viruses are important to understand viral pathogenesis and response to selective pressures. There is a need to characterize the complexity of mutant spectra in coronaviruses sampled from infected patients. In particular, the possible relationship between SARS-CoV-2 mutant spectrum complexity and disease associations has not been established. In the present study, we report an ultradeep sequencing (UDS) analysis of the mutant spectrum of amplicons from the nsp12 (polymerase)- and spike (S)-coding regions of 30 nasopharyngeal isolates (diagnostic samples) of SARS-CoV-2 of the first COVID-19 pandemic wave (Madrid, Spain, April 2020) classified according to the severity of ensuing COVID-19. Low-frequency mutations and deletions, counted relative to the consensus sequence of the corresponding isolate, were overwhelmingly abundant. We show that the average number of different point mutations, mutations per haplotype, and several diversity indices was significantly higher in SARS-CoV-2 isolated from patients who developed mild disease than in those associated with moderate or severe disease (exitus). No such bias was observed with RNA deletions. Location of amino acid substitutions in the three-dimensional structures of nsp12 (polymerase) and S suggest significant structural or functional effects. Thus, patients who develop mild symptoms may be a richer source of genetic variants of SARS-CoV-2 than patients with moderate or severe COVID-19. IMPORTANCE The study shows that mutant spectra of SARS-CoV-2 from diagnostic samples differ in point mutation abundance and complexity and that significantly larger values were observed in virus from patients who developed mild COVID-19 symptoms. Mutant spectrum complexity is not a uniform trait among isolates. The nature and location of low-frequency amino acid substitutions present in mutant spectra anticipate great potential for phenotypic diversification of SARS-CoV-2.
Collapse
Affiliation(s)
- Brenda Martínez-González
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - María Eugenia Soria
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Lucía Vázquez-Sirvent
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Cristina Ferrer-Orta
- Structural Biology Department, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
| | - Rebeca Lobo-Vega
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Pablo Mínguez
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Lorena de la Fuente
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Carlos Llorens
- Biotechvana, “Scientific Park”, Universidad de Valencia, Valencia, Spain
| | - Beatriz Soriano
- Biotechvana, “Scientific Park”, Universidad de Valencia, Valencia, Spain
| | - Ricardo Ramos
- Unidad de Genómica, “Scientific Park of Madrid”, Campus de Cantoblanco, Madrid, Spain
| | - Marta Cortón
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosario López-Rodríguez
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos García-Crespo
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Isabel Gallego
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Isabel de Ávila
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Jordi Gómez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Parasitología y Biomedicina ‘López-Neyra’ (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Llanos Salar-Vidal
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Jaime Esteban
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Ricardo Fernandez-Roblas
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Ignacio Gadea
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Carmen Ayuso
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Ruíz-Hornillos
- Allergy Unit, Hospital Infanta Elena, Valdemoro, Madrid, Spain
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
| | - Nuria Verdaguer
- Structural Biology Department, Institut de Biología Molecular de Barcelona CSIC, Barcelona, Spain
| | - Esteban Domingo
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Celia Perales
- Department of Clinical Microbiology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
40
|
Al-Qahtani AA. Mutations in the genome of severe acute respiratory syndrome coronavirus 2: implications for COVID-19 severity and progression. J Int Med Res 2022; 50:3000605221086433. [PMID: 35352580 PMCID: PMC8973081 DOI: 10.1177/03000605221086433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/22/2022] [Indexed: 01/02/2023] Open
Abstract
Coronaviridae is a large family of enveloped, positive-strand RNA viruses that has plagued the world since it was discovered in humans in the 1960s. The recent severe acute respiratory syndrome coronavirus (SARS-CoV)-2 pandemic has already exceeded the number of combined cases and deaths witnessed during previous SARS-CoV and Middle East respiratory syndrome-CoV epidemics in the last two decades. This narrative review focuses on genomic mutations in SARS-CoV-2 and their impact on the severity and progression of COVID-19 in light of reported data in the literature. Notable SARS-CoV-2 mutations associated with open reading frames, the S glycoprotein, and nucleocapsid protein, currently circulating globally, are discussed along with emerging mutations such as those in the SARS-CoV-2 VUI 202012/01 variant in the UK and other European countries, the 484K.V2 and P.1 variants in Brazil, the B.1.617 variant in India, and South African variants 501Y.V2 and B.1.1.529 (omicron). These variants have the potential to influence the receptor binding domain, host-virus fusion, and SARS-CoV-2 replication. Correlating these mutations with disease dynamics could help us understand their pathogenicity and design appropriate therapeutics.
Collapse
Affiliation(s)
- Ahmed Ali Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, Alfaisal University, School of Medicine, Riyadh, Saudi Arabia
| |
Collapse
|
41
|
Zhang Y, Wu G, Chen S, Ju X, Yimaer W, Zhang W, Lin S, Hao Y, Gu J, Li J. A review on COVID-19 transmission, epidemiological features, prevention and vaccination. MEDICAL REVIEW 2022; 2:23-49. [PMID: 35658107 PMCID: PMC9047653 DOI: 10.1515/mr-2021-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/13/2021] [Indexed: 11/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused hundreds of millions of infections and millions of deaths over past two years. Currently, many countries have still not been able to take the pandemic under control. In this review, we systematically summarized what we have done to mitigate the COVID-19 pandemic, from the perspectives of virus transmission, public health control measures, to the development and vaccination of COVID-19 vaccines. As a virus most likely coming from bats, the SARS-CoV-2 may transmit among people via airborne, faecal-oral, vertical or foodborne routes. Our meta-analysis suggested that the R0 of COVID-19 was 2.9 (95% CI: 2.7–3.1), and the estimates in Africa and Europe could be higher. The median Rt could decrease by 23–96% following the nonpharmacological interventions, including lockdown, isolation, social distance, and face mask, etc. Comprehensive intervention and lockdown were the most effective measures to control the pandemic. According to the pooled R0 in our meta-analysis, there should be at least 93.3% (95% CI: 89.9–96.2%) people being vaccinated around the world. Limited amount of vaccines and the inequity issues in vaccine allocation call for more international cooperation to achieve the anti-epidemic goals and vaccination fairness.
Collapse
Affiliation(s)
- Yuqin Zhang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Gonghua Wu
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Shirui Chen
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xu Ju
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | | | - Wangjian Zhang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Shao Lin
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, NY, USA
| | - Yuantao Hao
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
- Sun Yat-Sen University Global Health Institute, School of Public Health and Institute of State Governance, Sun Yat-Sen University, Guangzhou, China
| | - Jing Gu
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Jinghua Li
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
42
|
Petrenko VA, Gillespie JW, De Plano LM, Shokhen MA. Phage-Displayed Mimotopes of SARS-CoV-2 Spike Protein Targeted to Authentic and Alternative Cellular Receptors. Viruses 2022; 14:v14020384. [PMID: 35215976 PMCID: PMC8879608 DOI: 10.3390/v14020384] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
The evolution of the SARS-CoV-2 virus during the COVID-19 pandemic was accompanied by the emergence of new heavily mutated viral variants with increased infectivity and/or resistance to detection by the human immune system. To respond to the urgent need for advanced methods and materials to empower a better understanding of the mechanisms of virus’s adaptation to human host cells and to the immuno-resistant human population, we suggested using recombinant filamentous bacteriophages, displaying on their surface foreign peptides termed “mimotopes”, which mimic the structure of viral receptor-binding sites on the viral spike protein and can serve as molecular probes in the evaluation of molecular mechanisms of virus infectivity. In opposition to spike-binding antibodies that are commonly used in studying the interaction of the ACE2 receptor with SARS-CoV-2 variants in vitro, phage spike mimotopes targeted to other cellular receptors would allow discovery of their role in viral infection in vivo using cell culture, tissue, organs, or the whole organism. Phage mimotopes of the SARS-CoV-2 Spike S1 protein have been developed using a combination of phage display and molecular mimicry concepts, termed here “phage mimicry”, supported by bioinformatics methods. The key elements of the phage mimicry concept include: (1) preparation of a collection of p8-type (landscape) phages, which interact with authentic active receptors of live human cells, presumably mimicking the binding interactions of human coronaviruses such as SARS-CoV-2 and its variants; (2) discovery of closely related amino acid clusters with similar 3D structural motifs on the surface of natural ligands (FGF1 and NRP1), of the model receptor of interest FGFR and the S1 spike protein; and (3) an ELISA analysis of the interaction between candidate phage mimotopes with FGFR3 (a potential alternative receptor) in comparison with ACE2 (the authentic receptor).
Collapse
Affiliation(s)
- Valery A. Petrenko
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Correspondence: (V.A.P.); (J.W.G.); Tel.: +1-334-844-2897 (V.A.P.); +1-334-844-2625 (J.W.G.)
| | - James W. Gillespie
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Correspondence: (V.A.P.); (J.W.G.); Tel.: +1-334-844-2897 (V.A.P.); +1-334-844-2625 (J.W.G.)
| | - Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy;
| | | |
Collapse
|
43
|
Golshani M, Hrdý J. Multiple Sclerosis Patients and Disease Modifying Therapies: Impact on Immune Responses against COVID-19 and SARS-CoV-2 Vaccination. Vaccines (Basel) 2022; 10:vaccines10020279. [PMID: 35214735 PMCID: PMC8876554 DOI: 10.3390/vaccines10020279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
This article reviews the literature on SARS-CoV-2 pandemic and multiple sclerosis (MS). The first part of the paper focuses on the current data on immunopathology of SARS-CoV-2 and leading vaccines produced against COVID-19 infection. In the second part of the article, we discuss the effect of Disease Modifying Therapies (DMTs) on COVID-19 infection severity or SARS-CoV-2 vaccination in MS patients plus safety profile of different vaccine platforms in MS patients.
Collapse
Affiliation(s)
| | - Jiří Hrdý
- Correspondence: ; Tel.: +420-224968509
| |
Collapse
|
44
|
Sitjar J, Xu HZ, Liu CY, Wang JR, Liao JD, Tsai HP, Lee H, Liu BH, Chang CW. Synergistic surface-enhanced Raman scattering effect to distinguish live SARS-CoV-2 S pseudovirus. Anal Chim Acta 2022; 1193:339406. [PMID: 35058004 PMCID: PMC8711038 DOI: 10.1016/j.aca.2021.339406] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022]
Abstract
The COVID-19 pandemic negatively affected the economy and health security on a global scale, causing a drastic change on lifestyle, calling a need to mitigate further transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Surface-enhanced Raman spectroscopy (SERS) has shown great potential in the sensitive and rapid detection of various molecules including viruses, through the identification of characteristic peaks of their outer membrane proteins. Accurate detection can be developed through the synergistic integration effect among SERS-active substrate, the appropriate laser wavelength, and the target analyte. In this study, gold nanocavities (Au NC) and Au nanoparticles upon ZrO2 nano-bowls (Au NPs/pZrO2) were tested and used as SERS-active substrates in detecting SARS-CoV-2 pseudovirus containing S protein as a surface capsid glycoprotein (SARS-CoV-2 S pseudovirus) and vesicular stomatitis virus G (VSV-G) pseudo-type lentivirus (VSV-G pseudovirus) to demonstrate their virus detection capability. The optimized Au NCs and Au NPs/pZrO2 substrates were then verified by examining the repetition of measurement, reproducibility, and detection limit. Due to the difference in geometry and composition of the substrates, the characteristic peak-positions of live SARS-CoV-2 S and VSV-G pseudoviruses in the obtained Raman spectra vary, which were also compared with those of inactivated ones. Based on the experimental results, SERS mechanism of each substrate to detect virus is proposed. The formation of hot spots brought by the synergistic integration effect among substrate, analyte, and laser induction may result differences in the obtained SERS spectra.
Collapse
Affiliation(s)
- Jaya Sitjar
- Engineered Materials for Biomedical Applications Laboratory, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Hong-Zheng Xu
- Engineered Materials for Biomedical Applications Laboratory, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Chih-Yun Liu
- Engineered Materials for Biomedical Applications Laboratory, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan; Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Jiunn-Der Liao
- Engineered Materials for Biomedical Applications Laboratory, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Huey-Pin Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan; Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Han Lee
- Engineered Materials for Biomedical Applications Laboratory, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Bernard Haochih Liu
- Laboratory for Micro/Nanofabrication and Nanoanalysis, Department of Materials Science and Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Chia-Wei Chang
- MAN Technology Co. Ltd, 1F, No. 97, Yunong 3rd St., Tainan, 701, Taiwan.
| |
Collapse
|
45
|
Gusev E, Sarapultsev A, Solomatina L, Chereshnev V. SARS-CoV-2-Specific Immune Response and the Pathogenesis of COVID-19. Int J Mol Sci 2022; 23:1716. [PMID: 35163638 PMCID: PMC8835786 DOI: 10.3390/ijms23031716] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The review aims to consolidate research findings on the molecular mechanisms and virulence and pathogenicity characteristics of coronavirus disease (COVID-19) causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and their relevance to four typical stages in the development of acute viral infection. These four stages are invasion; primary blockade of antiviral innate immunity; engagement of the virus's protection mechanisms against the factors of adaptive immunity; and acute, long-term complications of COVID-19. The invasion stage entails the recognition of the spike protein (S) of SARS-CoV-2 target cell receptors, namely, the main receptor (angiotensin-converting enzyme 2, ACE2), its coreceptors, and potential alternative receptors. The presence of a diverse repertoire of receptors allows SARS-CoV-2 to infect various types of cells, including those not expressing ACE2. During the second stage, the majority of the polyfunctional structural, non-structural, and extra proteins SARS-CoV-2 synthesizes in infected cells are involved in the primary blockage of antiviral innate immunity. A high degree of redundancy and systemic action characterizing these pathogenic factors allows SARS-CoV-2 to overcome antiviral mechanisms at the initial stages of invasion. The third stage includes passive and active protection of the virus from factors of adaptive immunity, overcoming of the barrier function at the focus of inflammation, and generalization of SARS-CoV-2 in the body. The fourth stage is associated with the deployment of variants of acute and long-term complications of COVID-19. SARS-CoV-2's ability to induce autoimmune and autoinflammatory pathways of tissue invasion and development of both immunosuppressive and hyperergic mechanisms of systemic inflammation is critical at this stage of infection.
Collapse
Affiliation(s)
- Evgenii Gusev
- Laboratory of Immunology of Inflammation, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Alexey Sarapultsev
- Laboratory of Immunology of Inflammation, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Liliya Solomatina
- Laboratory of Immunology of Inflammation, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Valeriy Chereshnev
- Laboratory of Immunology of Inflammation, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| |
Collapse
|
46
|
Hao X, Peng X, Ding X, Qin Y, Lv M, Li J, Li K. Application of digital education in undergraduate nursing and medical interns during the COVID-19 pandemic: A systematic review. NURSE EDUCATION TODAY 2022; 108:105183. [PMID: 34741918 PMCID: PMC8545701 DOI: 10.1016/j.nedt.2021.105183] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/24/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Due to the rapid spread of coronavirus disease 2019 (COVID-19) around the world, the World Health Organization (WHO) declared it a global pandemic on March 11, 2020. This declaration had an unprecedented impact on health profession education, especially the clinical clerkship of nursing and medical students. The teaching hospitals had to suspend traditional bedside clinical teaching and switch to digital education. OBJECTIVE To systematically synthesize the available literature on the application of digital education in undergraduate nursing and medical interns during the COVID-19 pandemic. DESIGN A systematic review informed by PRISMA guidelines. DATA SOURCES Five electronic databases were systematically searched: PubMed, Embase, MEDLINE (OVID), CINAHL and the Cochrane Library. REVIEW METHODS The retrieved articles were screened at the title, abstract, and full text stages. The Mixed-Methods Appraisal Tool (MMAT) was used to assess the quality of quantitative and mixed-method studies. Then, two reviewers extracted the quantitative data of the included studies. RESULTS A total of 4596 studies were identified following a comprehensive search, and 16 studies were included after removing duplicates and screening, which focused on undergraduate nursing students (3 studies) and medical students (13 studies). We found that the standalone digital education modalities were as effective as conventional learning for knowledge and practice. Different educational technologies have different effects on the knowledge and practice of interns. CONCLUSION Digital education plays a significant role in distance training for nursing and medical interns both now and in the future. The overall risk of bias was high, and the quality of evidence was found to be variable. There is a need for further research designing more quasi-experimental studies to assess the effectiveness of standalone digital education interventions for the remote training of nursing or medical interns to be fully prepared for emergencies.
Collapse
Affiliation(s)
- Xiaonan Hao
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China
| | - Xin Peng
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China
| | - Xinxin Ding
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China.
| | - Yuan Qin
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China
| | - Miaohua Lv
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China
| | - Jing Li
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China.
| | - Kun Li
- School of Nursing, Jilin University, 965Xinjiang street, Changchun 130021, China.
| |
Collapse
|
47
|
Hilverda F, Vollmann M. The Role of Risk Perception in Students' COVID-19 Vaccine Uptake: A Longitudinal Study. Vaccines (Basel) 2021; 10:22. [PMID: 35062683 PMCID: PMC8777829 DOI: 10.3390/vaccines10010022] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/05/2022] Open
Abstract
Since COVID-19 vaccine uptake was found to be especially low among young adults, the present study investigated COVID-19 risk perception as predictor of COVID-19 vaccination intention and actual COVID-19 vaccine uptake among this age group. More specifically, it was tested whether cognitive risk perception predicts vaccination uptake successively via affective risk perception and vaccination intention. In total, 680 students (65.9% female) between 17 and 28 years participated in this longitudinal online study. COVID-19 cognitive and affective risk perception, COVID-19 vaccination intention, and actual COVID-19 vaccine uptake were measured in t1: November/December 2020, t2: March 2021, and t3: June/July 2021, respectively. The mediation analysis revealed a significant indirect effect of perceived severity at t1 on vaccine uptake at t3 via worry at t1 and vaccination intention at t2. Stronger perceptions of perceived severity of COVID-19 were related to more worry about COVID-19, which led to a higher vaccination intention, which, in turn, increased the chance of COVID-19 vaccine uptake. To increase vaccine uptake among young adults it might be fruitful to emphasize the severity of COVID-19. However, one should take into account that tapping into fear works best when messages also include efficacy statements.
Collapse
Affiliation(s)
- Femke Hilverda
- Department of Socio-Medical Sciences, Erasmus School of Health Policy & Management, Erasmus University Rotterdam, 3000 DR Rotterdam, The Netherlands;
| | | |
Collapse
|
48
|
Berar Yanay N, Freimann S, Shapira M, Gershkovich R, Peleg N, Mtanis L, Armaly Z. Antibody response in BNT162b2 mRNA-vaccinated dialysis patients and those with coronavirus disease 2019: Long-term follow-up. Hemodial Int 2021; 26:281-283. [PMID: 34939309 DOI: 10.1111/hdi.12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/01/2022]
Affiliation(s)
- Noa Berar Yanay
- Department of Nephrology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Sarit Freimann
- Laboratory Division, Hillel Yaffe Medical Center, Hadera, Israel
| | - Maanit Shapira
- Laboratory Division, Hillel Yaffe Medical Center, Hadera, Israel.,Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel
| | | | - Nana Peleg
- Department of Nephrology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Loai Mtanis
- Department of Nephrology and Hypertension, Nazareth Hospital, EMMS, Nazareth, Israel
| | - Zaher Armaly
- Department of Nephrology and Hypertension, Nazareth Hospital, EMMS, Nazareth, Israel.,Azrieli Faculty of Medicine in Safed, Safed, Israel
| |
Collapse
|
49
|
Najjar H, Al-Jighefee HT, Qush A, Ahmed MN, Awwad S, Kamareddine L. COVID-19 Vaccination: The Mainspring of Challenges and the Seed of Remonstrance. Vaccines (Basel) 2021; 9:1474. [PMID: 34960220 PMCID: PMC8707780 DOI: 10.3390/vaccines9121474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
As of March 2020, the time when the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became a pandemic, our existence has been threatened and the lives of millions have been claimed. With this ongoing global issue, vaccines are considered of paramount importance in curtailing the outbreak and probably a prime gamble to bring us back to 'ordinary life'. To date, more than 200 vaccine candidates have been produced, many of which were approved by the Food and Drug Administration (FDA) for emergency use, with the research and discovery phase of their production process passed over. Capering such a chief practice in COVID-19 vaccine development, and manufacturing vaccines at an unprecedented speed brought many challenges into play and raised COVID-19 vaccine remonstrance. In this review, we highlight relevant challenges to global COVID-19 vaccine development, dissemination, and deployment, particularly at the level of large-scale production and distribution. We also delineate public perception on COVID-19 vaccination and outline the main facets affecting people's willingness to get vaccinated.
Collapse
Affiliation(s)
- Hoda Najjar
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Hadeel T. Al-Jighefee
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Abeer Qush
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Muna Nizar Ahmed
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Sara Awwad
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
| | - Layla Kamareddine
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (H.N.); (H.T.A.-J.); (A.Q.); (M.N.A.); (S.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
50
|
Ortuño FM, Loucera C, Casimiro-Soriguer CS, Lepe JA, Camacho Martinez P, Merino Diaz L, de Salazar A, Chueca N, García F, Perez-Florido J, Dopazo J. Highly accurate whole-genome imputation of SARS-CoV-2 from partial or low-quality sequences. Gigascience 2021; 10:giab078. [PMID: 34865008 PMCID: PMC8643610 DOI: 10.1093/gigascience/giab078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The current SARS-CoV-2 pandemic has emphasized the utility of viral whole-genome sequencing in the surveillance and control of the pathogen. An unprecedented ongoing global initiative is producing hundreds of thousands of sequences worldwide. However, the complex circumstances in which viruses are sequenced, along with the demand of urgent results, causes a high rate of incomplete and, therefore, useless sequences. Viral sequences evolve in the context of a complex phylogeny and different positions along the genome are in linkage disequilibrium. Therefore, an imputation method would be able to predict missing positions from the available sequencing data. RESULTS We have developed the impuSARS application, which takes advantage of the enormous number of SARS-CoV-2 genomes available, using a reference panel containing 239,301 sequences, to produce missing data imputation in viral genomes. ImpuSARS was tested in a wide range of conditions (continuous fragments, amplicons or sparse individual positions missing), showing great fidelity when reconstructing the original sequences, recovering the lineage with a 100% precision for almost all the lineages, even in very poorly covered genomes (<20%). CONCLUSIONS Imputation can improve the pace of SARS-CoV-2 sequencing production by recovering many incomplete or low-quality sequences that would be otherwise discarded. ImpuSARS can be incorporated in any primary data processing pipeline for SARS-CoV-2 whole-genome sequencing.
Collapse
Affiliation(s)
- Francisco M Ortuño
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - Carlos Loucera
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - Carlos S Casimiro-Soriguer
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - Jose A Lepe
- Unidad Clínica Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Pedro Camacho Martinez
- Unidad Clínica Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Laura Merino Diaz
- Unidad Clínica Enfermedades Infecciosas, Microbiología y Medicina Preventiva, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
| | - Adolfo de Salazar
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Natalia Chueca
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Federico García
- Servicio de Microbiología, Hospital Universitario San Cecilio, 18016 Granada, Spain
| | - Javier Perez-Florido
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - Joaquin Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocio, 41013 Sevilla, Spain
- FPS/ELIXIR-es, Hospital Virgen del Rocío, Sevilla 42013, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Hospital Universitario San Cecilio, 18016 Granada, Spain
| |
Collapse
|