1
|
Li M, Wu Y, Tian J, Yang Q, Gao M, Wang Y, Wang X, Zhang J, Pan Y, Shi H, Shi D, Zhang X, Chen J, Guo L, Feng L. Nuclear shuttling of CDC4 mediated broad-spectrum antiviral activity against diverse coronaviruses. Emerg Microbes Infect 2025:2493922. [PMID: 40260685 DOI: 10.1080/22221751.2025.2493922] [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: 04/24/2025]
Abstract
Pandemics of coronavirus (CoV)-related infection have been a major issue since the outbreaks of SARS, MERS and COVID-2019 in the past decades, leading a substantial threat to public health. Porcine deltacoronavirus (PDCoV), a new swine coronavirus, causes enteropathogenic disease characterized by acute diarrhea, vomiting and dehydration in suckling piglets and poses potential risks of cross-species transmission. Here we reveal a novel function of CDC4 protein in restricting PDCoV infection. Ectopic expression of CDC4 suppresses PDCoV replication, whereas knockdown of CDC4 expression enhances PDCoV infection. Importantly, it was revealed that PDCoV encoded nucleocapsid (N) was involved in CDC4 nuclear-cytoplasmic shuttling, which was critical for CDC4 to exert the antiviral activity against PDCoV replication. Mechanistically, PDCoV N protein was detected to specifically interact with RIG-I to antagonize RIG-I-like receptor (RLR)-mediated IFN-β production, leading to disruptions of host innate immune defense. Meanwhile, CDC4 was proved to interact with PDCoV N protein and disrupted the interaction between PDCoV N and RIG-I, resulting in alleviated antagonism of IFN-β production mediated by PDCoV N. Similarly, a broad-spectrum inhibitory effects of CDC4 on N mediated antagonism were confirmed by the shared mechanisms among the different coronaviruses from Coronaviridae family, such as transmissible gastroenteritis virus (TGEV) from Alphacoronavirus (α-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from Betacoronavirus (β-CoV). Therefore, a novel antiviral role of CDC4 was elucidated that CDC4 competes binding with CoVs N proteins to suppress CoVs N mediated antagonism of RLR associated signaling pathway in the context of diverse coronavirus infections.
Collapse
Affiliation(s)
- Mingwei Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yang Wu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Qian Yang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Mingze Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongrui Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuepeng Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ju Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yudi Pan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Da Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jianfei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Longjun Guo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
2
|
Lertwanichwattana T, Srivanichakorn S, Noknoy S, Ratchaseema SSN, Phanuphak N, Wongthavarawat K, Siriussawakul A, Srinonprasert V, Leelahavarong P, Chevaisrakul P, Lumjiaktase P, Kumpitak A, Phromsri N, Sirisinsuk Y, Kietdumrongwong P, Aramrattana A, Rangsin R. Outcomes of Home Isolation Care Among COVID-19 Patients During the 2021 Epidemic Crisis in the Bangkok Metropolitan Region, Thailand. Am J Public Health 2025; 115:605-616. [PMID: 39883900 PMCID: PMC11903064 DOI: 10.2105/ajph.2024.307922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Objectives. To determine the overall mortality and risk factors of COVID-19 patients who were admitted to the Home Isolation (HI) program in Bangkok, Thailand, during the epidemic crisis in 2021. Methods. We conducted a retrospective cohort study using the data from a government telehealth application from July to December 2021. The vital status was verified from the government database on September 20, 2022. We used survival analysis to analyze the 28-day mortality and independently associated factors. Results. Of 90 854 reported cases, the average age was 37.27 years, and half were men. Initial symptoms included being asymptomatic (51.66%), having mild symptoms (35.60%), or experiencing severe symptoms requiring nonurgent (11.27%) or urgent referral (1.47%). The 28-day mortality rate was 0.80%. Factors associated with 28-day mortality included older age, male gender, higher body mass index, severity of initial symptoms, and time to admission. Conclusions. The Home Isolation program was able to manage a high volume of patients, including severe cases, exceeding its initial design. Thailand's COVID-19 mortality rate remained relatively low compared with other countries. Proactive bed surge planning and continuous plan improvement were crucial for future preparedness. (Am J Public Health. 2025;115(4):605-616. https://doi.org/10.2105/AJPH.2024.307922).
Collapse
Affiliation(s)
- Teeraboon Lertwanichwattana
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Supattra Srivanichakorn
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sairat Noknoy
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinapa Siriporn Na Ratchaseema
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nittaya Phanuphak
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Wongthavarawat
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Arunotai Siriussawakul
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Varalak Srinonprasert
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattara Leelahavarong
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Parawee Chevaisrakul
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Putthapoom Lumjiaktase
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Aree Kumpitak
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nopphan Phromsri
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Yupadee Sirisinsuk
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pongtorn Kietdumrongwong
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apinun Aramrattana
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Ram Rangsin
- Teeraboon Lertwanichwattana and Ram Rangsin are with Phramongkutklao College of Medicine, Bangkok, Thailand. Supattra Srivanichakorn, Sairat Noknoy, and Sirinapa Siriporn Na Ratchaseema are with the Royal College of Family Physicians of Thailand, Bangkok. Nittaya Phanuphak is with the Institute of HIV Research and Innovation, Bangkok. Kitti Wongthavarawat is with the National Science and Technology Development Agency, Bangkok. Arunotai Siriussawakul, Varalak Srinonprasert, and Pattara Leelahavarong are with the Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Parawee Chevaisrakul and Putthapoom Lumjiaktase are with the Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok. Aree Kumpitak is with the Thai Network of People Living With HIV, Bangkok. Nopphan Phromsri is with the Human Settlement Foundation, Bangkok. Yupadee Sirisinsuk is with the Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok. Pongtorn Kietdumrongwong is with the Bangkok Dusit Medical Services, Bangkok. Apinun Aramrattana is with the Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
3
|
Zeng Z, Yao L, Feng H, Wang Z, Jiang L, Wang H, Zhou C, Shang Y, Wang H, Shao H, Wen G, Luo Q. Genetic and pathogenic characteristics of a novel recombinant GI-19 infectious bronchitis virus strain isolated from northeastern China. Poult Sci 2025; 104:104985. [PMID: 40081171 PMCID: PMC11946749 DOI: 10.1016/j.psj.2025.104985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Since the 1980s, despite vaccination, the infectious bronchitis virus (IBV) infection rate in commercial broilers and layers in China has continued to rise significantly, causing substantial economic losses to the poultry industry. In this study, an IBV strain was isolated from a layer farm in northeast China and named CK/CH/LN/2302. The whole genome sequence analysis revealed that CK/CH/LN/2302 shared a high level of homology (96.41 %) with the GI-19 strain SC/SDL/19. The phylogenetic tree based on the S1 gene indicates that CK/CH/LN/2302 belongs to the GI-19 lineage. Notably, recombination analysis using RDP5 and SimPlot software suggested that the GI-19 strain and a 4/91-like strain likely contributed to four recombination events in the CK/CH/LN/2302 genome. Phylogenetic analysis of these four regions further supported this conclusion. Protein structure analysis revealed that most of the nonstructural protein 2 (nsp2), main protease (Mpro), S1, and 5a protein regions were replaced by sequences from the 4/91-like strain. After infecting 1-day-old SPF chickens, CK/CH/LN/2302 presented a mortality rate as high as 60 %. Higher viral loads were detected in tissues such as the larynx, trachea, lungs, duodenum, jejunum and kidneys, indicating the multitissue tropism of this strain. Neutralization assay results revealed that the serum from 28-day-old commercial chickens immunized with the H120 vaccine was unable to effectively neutralize CK/CH/LN/2302. Compared with the S1 subunit of H120, CK/CH/LN/2302 demonstrated conformational changes, particularly in the hypervariable regions (HVRs), which may facilitate immune evasion. The genetic characteristics and pathogenicity of CK/CH/LN/2302 highlight the ongoing evolution of GI-19 IBV strains in China, emphasizing the urgent need for appropriate control strategies.
Collapse
Affiliation(s)
- Zhe Zeng
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Lun Yao
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Helong Feng
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China; Shannan Tibetan Chicken Industry Research Institute, Shannan 856000 Tibet, China
| | - Zichen Wang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Liren Jiang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Haojie Wang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Chengli Zhou
- Liaoning Agricultural Development Service Center, Shenyang, China
| | - Yu Shang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Hongcai Wang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Huabin Shao
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China
| | - Guoyuan Wen
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China; Hubei Hongshan Laboratory, Wuhan, China.
| | - Qingping Luo
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Wuhan, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Wuhan, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
4
|
Dolgova NV, Qureshi M, Latimer M, Grishin A, Cygler M, Vogt LI, Cotelesage JJH, Sokaras D, Kroll T, Pickering IJ, George GN. Structural Changes at the Zinc Active Site of ACE2 on Binding the SARS-CoV-2 Spike Protein Receptor Binding Domain. Inorg Chem 2025; 64:3831-3841. [PMID: 39962897 DOI: 10.1021/acs.inorgchem.4c04974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The causative agent of Covid-19 is the SARS-CoV-2 virus. Initiation of cell entry by SARS-CoV-2 is critically dependent upon binding of the SARS-CoV-2 spike protein to angiotensin-converting enzyme 2 (ACE2, EC 3.4.17.23). The mechanism of binding of the SARS-CoV-2 spike receptor binding domain to ACE2 is among the most intensively studied infection mechanisms of any pathogen, including a very large number of structural studies. ACE2 is a membrane-associated zinc carboxypeptidase, comprising three domains, the protease domain, a neck domain, and a membrane-spanning α-helical domain. In addition to its role as a carboxypeptidase, ACE2 is also a chaperone for a Na+-amino acid cotransporter called B0AT1, and in the presence of B0AT1, full-length ACE2 forms dimers. Most studies to date related to Covid-19 have employed just the ACE2 protease domain and have neglected any possible roles of the Zn2+-containing ACE2 active site. We show here that ACE2, including the neck domain in addition to the protease domain (and in the absence of B0AT1), is dimeric and shows distinctive allostery in its catalytic activity. In contrast, the intensively studied protease domain is monomeric and shows no allostery. Binding of the spike receptor binding domain (RBD) to dimeric ACE2 eliminates its allostery. X-ray absorption spectroscopy of Zn2+ ACE2 shows distinctive changes in the active site structure upon binding of spike RBD but only in the dimeric form. Taken together, our results indicate that the Zn2+-containing active site exhibits a notable level of flexibility and that the dimeric form of ACE2, including both protease and neck domains, likely presents a superior model for the study of ACE2-spike interactions than the monomeric ACE2.
Collapse
Affiliation(s)
- Natalia V Dolgova
- Calibr─California Institute for Biomedical Research, Scripps Research, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Muhammad Qureshi
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Matthew Latimer
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Andrey Grishin
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Miroslaw Cygler
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Linda I Vogt
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Julien J H Cotelesage
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Ingrid J Pickering
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N George
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
5
|
Doğan EE, Rasulova N, Bayramova F, Hacisahinoğulları H, Yalın GY, Selçukbiricik ÖS, Gül N, Üzüm AK, Karşıdağ K, Satman İ. The effect of COVID-19 pandemic on new-onset adult diabetes and its one-year follow-up. Prim Care Diabetes 2025; 19:74-81. [PMID: 39675997 DOI: 10.1016/j.pcd.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024]
Abstract
AIMS Bidirectional detrimental relationships between COVID-19 infection and diabetes have been described globally. However, new-onset diabetes in adults and its follow-up during the pandemic have not been sufficiently investigated. In this study, new-onset autoimmune and type 2 diabetes cases during the pandemic were compared to those before the pandemic, and the clinical course of new-onset diabetes during the pandemic was examined. METHODS In this single-center retrospective cohort study, clinical and laboratory characteristics of new-onset diabetes patients before the pandemic (n = 161) and during the pandemic (n = 144) were evaluated between March 2018 and March 2022. RESULTS A 1.85-fold increase in new-onset adult diabetes cases was observed during the pandemic compared to pre-pandemic period (p = 0.010), while the proportion of autoimmune and type 2 diabetes (T2D) did not change. During the pandemic, there was a 6.2-fold increase in autoimmune diabetes presented with DKA (p = 0.003). Insulin was preferred 1.7 times more frequently as initial treatment during the pandemic (p = 0.014), and mean HbA1c (p = 0.003) and C-peptide (p = 0.010) were higher. Clinical and laboratory data did not differ between PCR (+) and PCR (-) patients. At one-year follow-up, while only HbA1c decreased in the autoimmune diabetes; in T2D group fasting glucose, HbA1c, C-peptide, and lipid profile were significantly improved. CONCLUSIONS The pandemic led to increased new-onset adult diabetes presented with DKA. However, clinical and laboratory features were similar between PCR positive and negative cases. PCR-confirmed COVID-19 may not adversely affect the medium-term clinical course of new diabetes in adults.
Collapse
Affiliation(s)
- Elif Ece Doğan
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey.
| | - Nubar Rasulova
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Fatima Bayramova
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Hülya Hacisahinoğulları
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Gülşah Yenidünya Yalın
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Özlem Soyluk Selçukbiricik
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Nurdan Gül
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Ayşe Kubat Üzüm
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Kubilay Karşıdağ
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - İlhan Satman
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Istanbul Faculty of Medicine, Istanbul University, Turkey
| |
Collapse
|
6
|
Liu R, Li J, Salena BJ, Li Y. Aptamer and DNAzyme Based Colorimetric Biosensors for Pathogen Detection. Angew Chem Int Ed Engl 2025; 64:e202418725. [PMID: 39551709 PMCID: PMC11753613 DOI: 10.1002/anie.202418725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
The detection of pathogens is critical for preventing and controlling health hazards across clinical, environmental, and food safety sectors. Functional nucleic acids (FNAs), such as aptamers and DNAzymes, have emerged as versatile molecular tools for pathogen detection due to their high specificity and affinity. This review focuses on the in vitro selection of FNAs for pathogens, with emphasis on the selection of aptamers for specific biomarkers and intact pathogens, including bacteria and viruses. Additionally, the selection of DNAzymes for bacterial detection is discussed. The integration of these FNAs into colorimetric biosensors has enabled the development of simple, cost-effective diagnostic platforms. Both non-catalytic and catalytic colorimetric biosensors are explored, including those based on gold nanoparticles, polydiacetylenes, protein enzymes, G-quadruplexes, and nanozymes. These biosensors offer visible detection through color changes, making them ideal for point-of-care diagnostics. The review concludes by highlighting current challenges and future perspectives for advancing FNA-based colorimetric biosensing technologies for pathogen detection.
Collapse
Affiliation(s)
- Rudi Liu
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Bruno J. Salena
- Department of MedicineMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical SciencesMichael G. DeGroote Institute of Infectious Disease ResearchSchool of Biomedical EngineeringBiointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| |
Collapse
|
7
|
Yegnaswamy S, C SK, Aldaais E. Conformational dynamics of the membrane protein of MERS-CoV in comparison with SARS-CoV-2 in ERGIC complex. J Biomol Struct Dyn 2025:1-15. [PMID: 39755960 DOI: 10.1080/07391102.2024.2437529] [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: 01/28/2024] [Accepted: 05/21/2024] [Indexed: 01/07/2025]
Abstract
The present study explores the conformational dynamics of the membrane protein of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) within the Endoplasmic Reticulum-Golgi Intermediate Compartment (ERGIC) complex using an all-atomistic molecular dynamics simulation approach. Significant structural changes were observed in the N-terminal, C-terminal, transmembrane, and beta-sheet sandwich domains of the MERS-CoV membrane protein. This study also highlights the structural similarities between the MERS-CoV and the SARS-CoV-2 membrane proteins, particularly in how both exhibit a distinct kink in the transmembrane helix caused by aromatic residue-lipid interactions. A structural expansion below the transmembrane and above the beta-sheet sandwich domain within the dimer was observed in all the M-proteins. This site on the beta-sheet sandwich domains near the C-terminal end could serve as a potential drug-binding site. Notably, a stable helical structure was identified in the C-terminal domain of the MERS-CoV membrane protein, whereas a proper secondary structural conformation was not observed in the SARS-CoV-2 membrane protein. Further, the SARS-CoV-2 membrane protein exhibited stronger binding to the lipid bilayer than the MERS-CoV, indicating its greater structural stability within the ERGIC complex. The structural similarity between the membrane protein of MERS-CoV and SARS-CoV-2 suggests the feasibility of employing a common inhibitor against these beta-coronaviruses. Furthermore, this analysis enhances our understanding of the membrane protein's interactions with proteins and lipids, paving the way for therapeutic developments against these viruses.
Collapse
Affiliation(s)
- Subha Yegnaswamy
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Selvaa Kumar C
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Ebtisam Aldaais
- College of Applied Medical Sciences, lmam Abdulrahman Bin Faisal University (lAU), Dammam, Saudi Arabia
| |
Collapse
|
8
|
Hassan AM, Mühlemann B, Al-Subhi TL, Rodon J, El-Kafrawy SA, Memish Z, Melchert J, Bleicker T, Mauno T, Perlman S, Zumla A, Jones TC, Müller MA, Corman VM, Drosten C, Azhar EI. Ongoing Evolution of Middle East Respiratory Syndrome Coronavirus, Saudi Arabia, 2023-2024. Emerg Infect Dis 2025; 31:57-65. [PMID: 39641462 DOI: 10.3201/eid3101.241030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) circulates in dromedary camels in the Arabian Peninsula and occasionally causes spillover infections in humans. MERS-CoV diversity is poorly understood because of the lack of sampling during the COVID-19 pandemic. We collected 558 swab samples from dromedary camels in Saudi Arabia during November 2023-January 2024. We found 39% were positive for MERS-CoV RNA by reverse transcription PCR. We sequenced 42 MERS-CoVs and 7 human 229E-related coronaviruses from camel swab samples by using high-throughput sequencing. Sequences from both viruses formed monophyletic clades apical to recently available genomes. MERS-CoV sequences were most similar to B5 lineage sequences and harbored unique genetic features, including novel amino acid polymorphisms in the spike protein. Further characterization will be required to understand their effects. MERS-CoV spillover into humans poses considerable public health concerns. Our findings indicate surveillance and phenotypic studies are needed to identify and monitor MERS-CoV pandemic potential.
Collapse
|
9
|
Shirouchi M, Sumikawa Y, Yoshioka‐Maeda K, Yamamoto‐Mitani N. How did home care nurses support COVID-19 patients in Japan? A qualitative study. Jpn J Nurs Sci 2025; 22:e12646. [PMID: 39777986 PMCID: PMC11707408 DOI: 10.1111/jjns.12646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
AIM Home care nurses support patients with COVID-19 with mild to moderate symptoms at home due to the lack of community-based support. Little is known about how nurses initiated and maintained support for patients with COVID-19. This study explored the experiences of home care nurses in supporting patients with COVID-19 at home. METHODS Using snowball sampling, 21 home care nurses participated in semi-structured interviews conducted either in person or online between September 2021 and February 2023. A qualitative study using a grounded theory approach was conducted. RESULTS Home care nurses faced a situation where "suffering patients are overflowing" due to inadequate support from public health centers and designated hospitals. Despite their anxiety, they acted as a "flexible safety net by quickly compensating for uncertain support systems" for patients, families, and local health workers. They "built tentative support systems immediately" and "updated tentative support systems, own knowledge, skills, and mindset." Home care nurses also "rushed to patient/family" to provide timely support and "avoided preventable death." CONCLUSIONS Home care nurses act as a flexible safety net, preventing patient deaths by quickly compensating for uncertain support systems before and after initiating care. Their efforts complemented the inadequacies of traditional infectious disease control systems, typically managed by public health centers. Strengthening secondary and tertiary prevention systems is essential for home care nurses to save lives while delivering individualized care during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Mana Shirouchi
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yuka Sumikawa
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Gerontological Home Care and Long‐Term Care Nursing, Division of Health Sciences and Nursing, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Kyoko Yoshioka‐Maeda
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Community Health Nursing/Public Health Nursing, Division of Health Sciences and Nursing, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Noriko Yamamoto‐Mitani
- Global Nursing Research Center, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Gerontological Home Care and Long‐Term Care Nursing, Division of Health Sciences and Nursing, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
10
|
Wang X, Xiong L, Zhu Y, Liu S, Zhao W, Wu X, Seydimemet M, Li L, Ding P, Lin X, Liu J, Wang X, Duan Z, Lu W, Suo Y, Cui M, Yue J, Jin R, Zheng M, Xu Y, Mei L, Hu H, Lu X. Covalent DNA-Encoded Library Workflow Drives Discovery of SARS-CoV-2 Nonstructural Protein Inhibitors. J Am Chem Soc 2024; 146:33983-33996. [PMID: 39574309 DOI: 10.1021/jacs.4c12992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
The COVID-19 pandemic, exacerbated by persistent viral mutations, underscored the urgent need for diverse inhibitors targeting multiple viral proteins. In this study, we utilized covalent DNA-encoded libraries to discover innovative triazine-based covalent inhibitors for the 3-chymotrypsin-like protease (3CLpro, Nsp5) and the papain-like protease (PLpro) domains of Nsp3, as well as novel non-nucleoside covalent inhibitors for the nonstructural protein 12 (Nsp12, RdRp). Optimization through molecular docking and medicinal chemistry led to the development of LU9, a nonpeptide 3CLpro inhibitor with an IC50 of 0.34 μM, and LU10, whose crystal structure showed a distinct binding mode within the 3CLpro active site. The X-ray cocrystal structure of SARS-CoV-2 PLpro in complex with XD5 uncovered a previously unexplored binding site adjacent to the catalytic pocket. Additionally, a non-nucleoside covalent Nsp12 inhibitor XJ5 achieved a potency of 0.12 μM following comprehensive structure-activity relationship analysis and optimization. Molecular dynamics revealed a potential binding mode. These compounds offer valuable chemical probes for target validation and represent promising candidates for the development of SARS-CoV-2 antiviral therapies.
Collapse
Affiliation(s)
- Xudong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Liwei Xiong
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Ying Zhu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Sixiu Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wenfeng Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Xinyuan Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Mengnisa Seydimemet
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Linjie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Peiqi Ding
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Xian Lin
- Suzhou Institute of Materia Medica, No. 108 Yuxin Road, Suzhou, Jiangsu 215123, P. R. China
| | - Jiaxiang Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Xuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Zhiqiang Duan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Weiwei Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Yanrui Suo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Mengqing Cui
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Jinfeng Yue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Rui Jin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Mingyue Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yechun Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Lianghe Mei
- Suzhou Institute of Materia Medica, No. 108 Yuxin Road, Suzhou, Jiangsu 215123, P. R. China
| | - Hangchen Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Xiaojie Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
11
|
Simon AY, Badmalia MD, Paquette SJ, Manalaysay J, Czekay D, Kandel BS, Sultana A, Lung O, Babuadze GG, Shahhosseini N. Evolutionary Relationships of Unclassified Coronaviruses in Canadian Bat Species. Viruses 2024; 16:1878. [PMID: 39772188 PMCID: PMC11680298 DOI: 10.3390/v16121878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 01/11/2025] Open
Abstract
Bats are recognized as natural reservoirs for an array of diverse viruses, particularly coronaviruses, which have been linked to major human diseases like SARS-CoV and MERS-CoV. These viruses are believed to have originated in bats, highlighting their role in virus ecology and evolution. Our study focuses on the molecular characterization of bat-derived coronaviruses (CoVs) in Canada. Tissue samples from 500 bat specimens collected in Canada were analyzed using pan-coronavirus RT-PCR assays to detect the presence of CoVs from four genera: Alpha-CoVs, Beta-CoV, Gamma-CoV, and Delta-CoV. Phylogenetic analysis was performed targeting the RNA-dependent RNA polymerase (RdRP) gene. Our results showed an overall 1.4% CoV positivity rate in our bat sample size. Phylogenetic analysis based on the ~600 bp sequences led to the identification of an unclassified subgenus of Alpha-CoV, provisionally named Eptacovirus. The findings contribute to a better understanding of the diversity and evolution of CoVs found in the bat species of Canada. The current study underscores the significance of bats in the epidemiology of CoVs and enhances the knowledge of their genetic diversity and potential impact on global public health.
Collapse
Affiliation(s)
- Ayo Yila Simon
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Maulik D. Badmalia
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Sarah-Jo Paquette
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Jessica Manalaysay
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
- Departments of Chemistry & Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Dominic Czekay
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Bishnu Sharma Kandel
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
| | - Asma Sultana
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (A.S.); (O.L.)
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (A.S.); (O.L.)
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - George Giorgi Babuadze
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Nariman Shahhosseini
- Centre for Vector-Borne Diseases, National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada; (A.Y.S.); (M.D.B.); (S.-J.P.); (J.M.); (D.C.); (B.S.K.)
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
12
|
Hemnani M, da Silva PG, Thompson G, Poeta P, Rebelo H, Mesquita JR. Detection and Prevalence of Coronaviruses in European Bats: A Systematic Review. ECOHEALTH 2024; 21:125-140. [PMID: 39580592 PMCID: PMC11649736 DOI: 10.1007/s10393-024-01688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 05/23/2024] [Accepted: 07/15/2024] [Indexed: 11/25/2024]
Abstract
Bats are known hosts for a wide range of coronaviruses (CoVs), including those that cause severe acute respiratory syndrome (SARS-CoV-1) and Middle East respiratory syndrome (MERS-CoV). With the emergence of the COVID-19 pandemic caused by the SARS-CoV-2 virus, it has become increasingly important to understand the diversity and prevalence of CoVs in bat populations. This systematic review aimed to compile studies that have sampled CoVs from bats across Europe and assessed various aspects related to the testing of bat samples, including the country where the bats were collected, the CoV genomic region studied, the CoV genera that were detected, and the identification of bat species that were found to be carrying CoV. We identified 30 studies that assessed CoVs presence in bats across multiple countries including Italy, Germany, and various other nations with one or two studies each, which tested them for CoVs using a variety of matrices. CoVs were found in nine genera of bats, and the genomic regions included RdRp, ORF1a gene, as well as full genome, detecting α- and/or β-CoVs, with most of them being detectable only in faeces. This review provides a comprehensive overview of the CoVs detected in bats across Europe and highlights the importance of continued surveillance and monitoring of bat populations for potential emerging zoonotic CoVs.
Collapse
Affiliation(s)
- Mahima Hemnani
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
| | - Priscilla Gomes da Silva
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Gertrude Thompson
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal
- CIBIO/InBIO, BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os Montes e Alto Douro, 5000-801, Vila Real, Portugal
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 1099-085, Caparica, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os Montes e Alto Douro, 5000-801, Vila Real, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Vila Real, Portugal
| | - Hugo Rebelo
- CIBIO/InBIO, BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
| | - João R Mesquita
- ICBAS-School of Medicine and Biomedical Sciences, Porto University, 4050-313, Porto, Portugal.
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, Porto, Portugal.
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal.
| |
Collapse
|
13
|
Zhang Y, Tian C, Yu X, Yu G, Han X, Wang Y, Zhou H, Zhang S, Li M, Yang T, Sun Y, Tai W, Yin Q, Zhao G. Lung-Selective Delivery of mRNA-Encoding Anti-MERS-CoV Nanobody Exhibits Neutralizing Activity Both In Vitro and In Vivo. Vaccines (Basel) 2024; 12:1315. [PMID: 39771977 PMCID: PMC11680347 DOI: 10.3390/vaccines12121315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a highly pathogenic virus causing severe respiratory illness, with limited treatment options that are mostly supportive. The success of mRNA technology in COVID-19 vaccines has opened avenues for antibody development against MERS-CoV. mRNA-based antibodies, expressed in vivo, offer rapid adaptability to viral mutations while minimizing long-term side effects. This study aimed to develop a lung-targeted lipid nanoparticle (LNP) system for mRNA-encoding neutralizing nanobodies against MERS-CoV, proposing a novel therapeutic strategy. Methods: An mRNA-encoding nanobody NbMS10 (mRNA-NbMS10) was engineered for enhanced stability and reduced immunogenicity. This mRNA was encapsulated in lung-selective LNPs using microfluidics to form the LNP-mRNA-NbMS10 system. Efficacy was assessed through in vitro assays and in vivo mouse studies, focusing on antigen-binding, neutralization, and sustained nanobody expression in lung tissues. Results: The LNP-mRNA-NbMS10 system expressed the nanobody in vitro, showing strong antigen-binding and significant MERS-CoV pseudovirus neutralization. In vivo studies confirmed selective lung mRNA delivery, with high nanobody expression sustained for up to 24 h, confirming lung specificity and prolonged antiviral activity. Conclusions: Extensive in vitro and in vivo evaluations demonstrate the LNP-mRNA-NbMS10 system's potential as a scalable, cost-effective, and adaptable alternative to current MERS-CoV therapies. This innovative platform offers a promising solution for preventing and treating respiratory infections, and countering emerging viral threats.
Collapse
Affiliation(s)
- Yuhang Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China;
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030031, China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China; (X.Y.); (G.Y.)
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China; (X.Y.); (G.Y.)
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Haisheng Zhou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
| | - Shuai Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China;
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Tiantian Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Yali Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China;
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Guangyu Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
14
|
Huang Z, Wang Z, Liu Y, Ke C, Feng J, He B, Jiang T. The links between dietary diversity and RNA virus diversity harbored by the great evening bat (Ia io). MICROBIOME 2024; 12:246. [PMID: 39578858 PMCID: PMC11585108 DOI: 10.1186/s40168-024-01950-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Predator‒prey interactions and their dynamic changes provide frequent opportunities for viruses to spread among organisms and thus affect their virus diversity. However, the connections between dietary diversity and virus diversity in predators have seldom been studied. The avivorous bats, Ia io, show a seasonal pattern of dietary diversity. Although most of them primarily prey on insects in summer, they mainly prey on nocturnally migrating birds in spring and autumn. RESULTS In this study, we characterized the RNA virome of three populations of I. io in Southwest China during summer and autumn using viral metatranscriptomic sequencing. We also investigated the relationships between dietary diversity and RNA virus diversity by integrating DNA metabarcoding and viral metatranscriptomic sequencing techniques at the population level of I. io. We found 55 known genera belonging to 35 known families of RNA viruses. Besides detecting mammal-related viruses, which are the usual concern, we also found a high abundance of insect-related viruses and some bird-related viruses. We found that insect-related viruses were more abundant in summer, while the bird-related viruses were predominantly detected in autumn, which might be caused by the seasonal differences in prey selection by I. io. Additionally, a significant positive correlation was identified between prey diversity and total virus diversity. The more similar the prey composition, the more similar the total virus composition and the higher the count of potential new viruses. We also found that the relative abundance of Picornaviridae increased with increasing prey diversity and body mass. CONCLUSIONS In this study, significant links were found between RNA virus diversity and dietary diversity of I. io. The results implied that dynamic changes in predator-prey interactions may facilitate frequent opportunities for viruses to spread among organisms. Video Abstract.
Collapse
Affiliation(s)
- Zhenglanyi Huang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, 5268 Renmin Avenue, Changchun, 130024, China
| | - Zhiqiang Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, 5268 Renmin Avenue, Changchun, 130024, China
| | - Yingying Liu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, 5268 Renmin Avenue, Changchun, 130024, China
| | - Can Ke
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, 5268 Renmin Avenue, Changchun, 130024, China
| | - Jiang Feng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China.
- College of Life Science, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - Biao He
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, China.
- Key Laboratory of Vegetation Ecology of Education Ministry, Institute of Grassland Science, Northeast Normal University, 5268 Renmin Avenue, Changchun, 130024, China.
| |
Collapse
|
15
|
Yu J, Ge S, Li J, Zhang Y, Xu J, Wang Y, Liu S, Yu X, Wang Z. Interaction between coronaviruses and the autophagic response. Front Cell Infect Microbiol 2024; 14:1457617. [PMID: 39650836 PMCID: PMC11621220 DOI: 10.3389/fcimb.2024.1457617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
In recent years, the emergence and widespread dissemination of the coronavirus SARS-CoV-2 has posed a significant threat to global public health and social development. In order to safely and effectively prevent and control the spread of coronavirus diseases, a profound understanding of virus-host interactions is paramount. Cellular autophagy, a process that safeguards cells by maintaining cellular homeostasis under diverse stress conditions. Xenophagy, specifically, can selectively degrade intracellular pathogens, such as bacteria, fungi, viruses, and parasites, thus establishing a robust defense mechanism against such intruders. Coronaviruses have the ability to induce autophagy, and they manipulate this pathway to ensure their efficient replication. While progress has been made in elucidating the intricate relationship between coronaviruses and autophagy, a comprehensive summary of how autophagy either benefits or hinders viral replication remains elusive. In this review, we delve into the mechanisms that govern how different coronaviruses regulate autophagy. We also provide an in-depth analysis of virus-host interactions, particularly focusing on the latest data pertaining to SARS-CoV-2. Our aim is to lay a theoretical foundation for the development of novel coronavirus vaccines and the screening of potential drug targets.
Collapse
Affiliation(s)
- Jiarong Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shengqiang Ge
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Jinming Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | | | - Jiao Xu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yingli Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shan Liu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Xiaojing Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, Qingdao, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
16
|
Hoffmann M, Kleine-Weber H, Graichen L, Nehlmeier I, Kempf A, Moldenhauer AS, Braun E, Assiri AM, Kirchhoff F, Sauter D, Alkharsah KR, Pöhlmann S. Acquisition of a multibasic cleavage site does not increase MERS-CoV entry into Calu-3 human lung cells. J Virol 2024; 98:e0130524. [PMID: 39470207 PMCID: PMC11575293 DOI: 10.1128/jvi.01305-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024] Open
Abstract
Human-to-human transmission of the highly pathogenic Middle East respiratory syndrome coronavirus (MERS-CoV) is currently inefficient. However, there is concern that the virus might mutate and thereby increase its transmissibility and thus pandemic potential. The pandemic SARS-CoV-2 depends on a highly cleavable furin motif at the S1/S2 site of the viral spike (S) protein for efficient lung cell entry, transmission, and pathogenicity. Here, by employing pseudotyped particles, we investigated whether augmented cleavage at the S1/S2 site also increases MERS-CoV entry into Calu-3 human lung cells. We report that polymorphism T746K at the S1/S2 cleavage site or optimization of the furin motif increases S protein cleavage but not lung cell entry. These findings suggest that, unlike what has been reported for SARS-CoV-2, a highly cleavable S1/S2 site might not augment MERS-CoV infectivity for human lung cells.IMPORTANCEThe highly cleavable furin motif in the spike protein is required for robust lung cell entry, transmission, and pathogenicity of SARS-CoV-2. In contrast, it is unknown whether optimization of the furin motif in the spike protein of the pre-pandemic MERS-CoV increases lung cell entry and allows for robust human-human transmission. The present study indicates that this might not be the case. Thus, neither a naturally occurring polymorphism that increased MERS-CoV spike protein cleavage nor artificial optimization of the cleavage site allowed for increased spike-protein-driven entry into Calu-3 human lung cells.
Collapse
Affiliation(s)
- Markus Hoffmann
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Hannah Kleine-Weber
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Anna-Sophie Moldenhauer
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
| | - Elisabeth Braun
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Daniel Sauter
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Khaled R Alkharsah
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University (IAU), Dammam, Saudi Arabia
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center- Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| |
Collapse
|
17
|
Hassan AM, Bajrai LH, Alharbi AS, Alhamdan MM, Dwivedi VD, Azhar EI. Elucidating the role of PPARG inhibition in enhancing MERS virus immune response: A network pharmacology and computational drug discovery. J Infect Public Health 2024; 17:102561. [PMID: 39437592 DOI: 10.1016/j.jiph.2024.102561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Middle East Respiratory Syndrome (MERS) has become a severe zoonotic disease, posing significant public health concerns due to the lack of specific medications. This urgently demands the development of novel therapeutic molecules. Understanding MERS's genetic underpinnings and potential therapeutic targets is crucial for developing effective treatments. METHODS Two gene expression datasets (GSE81909 and GSE100504) were analyzed to identify differentially expressed genes (DEGs) using GEO2R. Furthermore, gene ontology (GO), pathway enrichment analysis, and protein-protein interaction (PPI) network were performed to understand the gene's functions. A possible drug target was identified, and an FDA-approved drug library was screened against the selected target using molecular docking and validated the findings through molecular dynamics simulation, principal component analysis, free energy landscape, and MM/GBSA calculations. RESULTS The study on GSE81909 and GSE100504 datasets with icMERS and MOCK samples at 24 and 48 h revealed an upregulation in 73 and 267 DEGs, respectively. In the network pharmacology, STAT1, MX1, DDX58, EIF2AK2, ISG15, IFIT1, IFIH1, OAS1, IRF9, and OASL were identified as the top 10 hub genes. STAT1 was identified as the most connected hub gene among these top 10 hub genes, which plays a crucial role in the immune response to the MERS virus. Further study on STAT1 showed that PPARG helps reduce STAT1, which could modulate the immune response. Therefore, by inhibiting PPARG, the immunological response can be successfully enhanced. The known inhibitor of PPARG, 570 (Farglitazar), was used as a control. Further, screening using Tanimoto and K-mean clustering was performed, from which three compounds were identified: 2267, 3478, and 40326. Compound 3478 showed characteristics similar to the control, indicating robust binding to PPARG. 3478 showed the highest negative binding free energy with -41.20 kcal/mol, indicating strong binding with PPARG. CONCLUSIONS These findings suggest that 3478 promises to be a potential inhibitor of PPARG, and further experimental investigations can explore its potential as a MERS inhibitor.
Collapse
Affiliation(s)
- Ahmed M Hassan
- Special Infectious Agents Unit - BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Leena H Bajrai
- Special Infectious Agents Unit - BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Azzah S Alharbi
- Special Infectious Agents Unit - BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Meshari M Alhamdan
- Special Infectious Agents Unit - BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; Family Medicine Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Bioinformatics Research Division, Quanta Calculus, Greater Noida, India.
| | - Esam I Azhar
- Special Infectious Agents Unit - BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia.
| |
Collapse
|
18
|
Althobaity YM, Alkhudaydi MH, Hill EM, Thompson RN, Tildesley MJ. The time between symptom onset and various clinical outcomes: a statistical analysis of MERS-CoV patients in Saudi Arabia. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240094. [PMID: 39569349 PMCID: PMC11576103 DOI: 10.1098/rsos.240094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/04/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024]
Abstract
In this study, we investigate the impact of demographic characteristics on Middle East respiratory syndrome coronavirus (MERS-CoV) cases in Saudi Arabia, specifically focusing on the time intervals between symptom onset and key events such as hospitalization, case confirmation, reporting and death. We estimate these intervals using data from 2196 cases occurring between June 2012 and January 2020, partitioning the data into four age groups (0-24 years, 25-49 years, 50-74 years and 75-100 years). The duration from symptom onset to hospitalization varies between age cohorts, ranging from 4.03 to 4.75 days, with the 75-100 age group experiencing the longest delay. The interval from symptom onset to case confirmation spans 5.83-8.24 days, and again, the 75-100 age group faces the lengthiest delay. The interval from symptom onset and case reporting ranges from 7.0 to 9.8 days, with the 75-100 age group experiencing the longest delay. The period from symptom onset to death varies across age groups (12.3-16.1 days), with elevated mortality rates during outbreaks. Importantly, we observe age-based differences in the risk of hospitalization and other measures of infection severity, including the probability of death conditional on hospitalization. Careful quantification of epidemiological characteristics, including inference of key epidemiological periods and assessments of differences between cases of different ages, plays a crucial role in understanding the progression of MERS-CoV outbreaks and formulating effective public health strategies to mitigate their impact.
Collapse
Affiliation(s)
| | | | - Edward M Hill
- The Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, School of Life Sciences and Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK
| | - Robin N Thompson
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, UK
| | - Michael J Tildesley
- The Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, School of Life Sciences and Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
19
|
Okeke KI, Ahamefule CS, Nnabuife OO, Orabueze IN, Iroegbu CU, Egbe KA, Ike AC. Antiseptics: An expeditious third force in the prevention and management of coronavirus diseases. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100293. [PMID: 39497935 PMCID: PMC11532748 DOI: 10.1016/j.crmicr.2024.100293] [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] [Indexed: 11/07/2024] Open
Abstract
Notably, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and coronavirus disease 2019 (COVID-19) have all had significant negative impact on global health and economy. COVID-19 alone, has resulted to millions of deaths with new cases and mortality still being reported in its various waves. The development and use of vaccines have not stopped the transmission of SARS coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, even among vaccinated individuals. The use of vaccines and curative drugs should be supplemented with adoption of simple hygiene preventive measures in the fight against the spread of the virus, especially for healthcare workers. Several virucidal topical antiseptics, such as povidone-iodine (PVP-I), citrox, cyclodextrins among others, have been demonstrated to be efficacious in the inactivation of SARS-CoV-2 and other coronaviruses in both in vitro and in vivo studies. The strategic application of these virucidal formulations could provide the additional impetus needed to effectively control the spread of the virus. We have here presented a simple dimension towards curtailing the dissemination of COVID-19, and other coronaviruses, through the application of effective oral, nasal and eye antiseptics among patients and medical personnel. We have further discussed the mechanism of action of some of these commonly available virucidal solutions while also highlighting some essential controversies in their use.
Collapse
Affiliation(s)
- Kizito I. Okeke
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Chukwuemeka Samson Ahamefule
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Obianuju O. Nnabuife
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Ibuchukwu N. Orabueze
- Department of Medical Microbiology, University of Nigeria Teaching Hospital Enugu, Enugu State, Nigeria
| | - Christian U. Iroegbu
- Department of Microbiology, Cross River University of Technology, Calabar, Cross River State, Nigeria
| | - Kingsley A. Egbe
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| | - Anthony C. Ike
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001 Enugu State, Nigeria
| |
Collapse
|
20
|
Seo H, Jang Y. Cold-adapted live attenuated MERS-CoV vaccine strain remains attenuated in mice after multiple passages in Vero cells at 37 °C. Arch Microbiol 2024; 206:393. [PMID: 39240318 DOI: 10.1007/s00203-024-04120-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/18/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic disease affecting camels and humans. The live attenuated vaccine represents a candidate human vaccine because it can induce strong immune responses in immunized hosts. The attenuated vaccine strain of the highly pathogenic virus can also be used to produce a cell-based vaccine in the BSL2 GMP facility. In this study, we evaluated the reversion potential of pathogenicity to pathogenic wild-type virus to ensure the safety of the live attenuated vaccine strain. We passaged our previously developed cold-adapted live attenuated MERS-CoV vaccine strain at 22 °C (EMC2012-CA22°C) in Vero cells at 37 °C as often as 15 times to determine the potential of pathogenicity reversion in hDPP4 (human dipeptidyl peptidase 4)-transgenic mice, K18-hDPP4. The serial passage of EMC2012-CA22°C in Vero cells at 37 °C up to 15 times did not result in pathogenicity reversion to wild-type MERS-CoV. In K18-hDPP4 mice infected with this virus, no weight loss or mortality was observed, and no virus was detected in tissues such as the lung, kidney, brain, and nasal turbinate. In addition, mice immunized with this virus produced a robust neutralizing antibody response and were fully protected from lethal challenge with wild-type MERS-CoV. The cold-adapted attenuated MERS-CoV vaccine strain (EMC2012-CA22°C) was not reverted to wild-type pathogenic virus after 15 passages in Vero cells at 37 °C.
Collapse
Affiliation(s)
- Heejeong Seo
- PioneerVaccine, Inc, Chungnam National University, 99 Dae-Hak Ro, Yuseong Gu, Daejeon, 34134, Republic of Korea
- College of Veterinary Medicine, Kyunpook National University, Daegu, 34134, Republic of Korea
| | - Yunyueng Jang
- PioneerVaccine, Inc, Chungnam National University, 99 Dae-Hak Ro, Yuseong Gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
21
|
Chandran D, Chakraborty S, Chandran D, Subedi D, Jisha AI, Chopra H, Rabaan AA, Al-Tawfiq JA, Islam MR, Dhama K. Middle East Respiratory Syndrome Coronavirus Could be a Priority Pathogen to Cause Public Health Emergency: Noticeable Features and Counteractive Measures. ENVIRONMENTAL HEALTH INSIGHTS 2024; 18:11786302241271545. [PMID: 39156879 PMCID: PMC11329908 DOI: 10.1177/11786302241271545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024]
Abstract
Middle East respiratory syndrome (MERS) is caused by a specific strain of the 6 types of human coronaviruses (HCoV). MERS-CoV has spread unchecked since it was first discovered in Saudi Arabia in 2012. The virus most likely spreads through nosocomial and zoonotic channels. Genetic analyses suggest that bats were the initial hosts and that the disease spread to camels. Person-to-person transmission occurs with varying frequency, being most prevalent in clinical settings and the least common among the general population and among close relatives. Due to the severity of the illness, high fatality rate, potential for epidemic spread, and lack of adequate medical countermeasures, the World Health Organization (WHO) continues to list MERS-CoV as a priority pathogen. While no specific antiviral medicines exist, a combination of antivirals has shown promise in recent clinical trials. Vaccines against MERS-CoV are critically needed and are currently being developed. Early diagnosis and implementing appropriate infection control measures are keys to preventing hospital-associated outbreaks. Preventive measures include avoiding raw or undercooked meats and other animal products, ensuring proper hand hygiene in healthcare settings and around dromedaries, educating the public and healthcare personnel about the disease, and adhering to other recommended practices. Countries with a high prevalence of MERS should adhere to regulations designed to limit the transmission of the virus. The recent spread of MERS-CoV highlights the importance of public awareness regarding the significance of reporting symptoms so that appropriate control measures can be adopted. The narrative review discusses the incidence of MERS, its clinical presentation, potential transmission routes, recent reports, preventative and control measures, and current therapeutic options.
Collapse
Affiliation(s)
- Deepak Chandran
- Department of Animal Husbandry, Government of Kerala, Kerala, India
| | - Sandip Chakraborty
- State Disease Investigation Laboratory, ARDD, Abhoynagar, Agartala, West Tripura, India
| | - Diljith Chandran
- KVG Dental College & Hospital, Kurunjibhag, Sullia, Karnataka, India
| | - Deepak Subedi
- Department of Poultry Science, University of Georgia, Athens, Georgia, USA
| | - Ankitha Indu Jisha
- Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, Tamil Nadu, India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - 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
| | - Jaffar A Al-Tawfiq
- Specialty Internal Medicine and Quality Patient Safety Department, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Infectious Diseases Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Infectious Diseases Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Md. Rabiul Islam
- School of Pharmacy, BRAC University, Merul Badda, Dhaka, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, India
| |
Collapse
|
22
|
Dai X, Xu R, Li N. The Interplay between Airway Cilia and Coronavirus Infection, Implications for Prevention and Control of Airway Viral Infections. Cells 2024; 13:1353. [PMID: 39195243 PMCID: PMC11353096 DOI: 10.3390/cells13161353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Coronaviruses (CoVs) are a class of respiratory viruses with the potential to cause severe respiratory diseases by infecting cells of the upper respiratory tract, bronchial epithelium, and lung. The airway cilia are distributed on the surface of respiratory epithelial cells, forming the first point of contact between the host and the inhaled coronaviruses. The function of the airway cilia is to oscillate and sense, thereby defending against and removing pathogens to maintain the cleanliness and patency of the respiratory tract. Following infection of the respiratory tract, coronaviruses exploit the cilia to invade and replicate in epithelial cells while also damaging the cilia to facilitate the spread and exacerbation of respiratory diseases. It is therefore imperative to investigate the interactions between coronaviruses and respiratory cilia, as well as to elucidate the functional mechanism of respiratory cilia following coronavirus invasion, in order to develop effective strategies for the prevention and treatment of respiratory viral infections. This review commences with an overview of the fundamental characteristics of airway cilia, and then, based on the interplay between airway cilia and coronavirus infection, we propose that ciliary protection and restoration may represent potential therapeutic approaches in emerging and re-emerging coronavirus pandemics.
Collapse
Affiliation(s)
| | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| |
Collapse
|
23
|
Jia R, Li Z, Hu S, Chang H, Zeng M, Liu P, Lu L, Xu M, Zhai X, Qian M, Xu J. Immunological characterization and comparison of children with COVID-19 from their adult counterparts at single-cell resolution. Front Immunol 2024; 15:1358725. [PMID: 39148728 PMCID: PMC11325098 DOI: 10.3389/fimmu.2024.1358725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction The immunological characteristics that could protect children with coronavirus disease 2019 (COVID-19) from severe or fatal illnesses have not been fully understood yet. Methods Here, we performed single-cell RNA sequencing (scRNA-seq) analysis on peripheral blood samples of 15 children (8 with COVID-19) and compared them to 18 adults (13 with COVID-19). Results The child-adult integrated single cell data indicated that children with the disease presented a restrained response to type I interferon in most of the major immune cell types, along with suppression of upstream interferon regulatory factor and toll-like receptor expression in monocytes, which was confirmed by in vitro interferon stimulation assays. Unlike adult patients, children with COVID-19 showed lower frequencies of activated proinflammatory CD14+ monocytes, possibly explaining the rareness of cytokine storm in them. Notably, natural killer (NK) cells in pediatric patients displayed potent cytotoxicity with a rich expression of cytotoxic molecules and upregulated cytotoxic pathways, whereas the cellular senescence, along with the Notch signaling pathway, was significantly downregulated in NK cells, all suggesting more robust cytotoxicity in NK cells of children than adult patients that was further confirmed by CD107a degranulation assays. Lastly, a modest adaptive immune response was evident with more naïve T cells but less activated and proliferated T cells while less naïve B cells but more activated B cells in children over adult patients. Conclusion Conclusively, this preliminary study revealed distinct cell frequency and activation status of major immune cell types, particularly more robust NK cell cytotoxicity in PBMC that might help protect children from severe COVID-19.
Collapse
Affiliation(s)
- Ran Jia
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Zifeng Li
- Department of Hematology and Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Shiwen Hu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Hailing Chang
- Department of Infectious Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Mei Zeng
- Department of Infectious Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Pengcheng Liu
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Lijuan Lu
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Menghua Xu
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xiaowen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, Children's Hospital of Fudan University, National Children's Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Clinical Laboratory, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Alatawi A, Gumel AB. Mathematical assessment of control strategies against the spread of MERS-CoV in humans and camels in Saudi Arabia. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:6425-6470. [PMID: 39176403 DOI: 10.3934/mbe.2024281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
A new mathematical model for the transmission dynamics and control of the Middle Eastern respiratory syndrome (MERS), a respiratory virus caused by MERS-CoV coronavirus (and primarily spread to humans by dromedary camels) that first emerged out of the Kingdom of Saudi Arabia (KSA) in 2012, was designed and used to study the transmission dynamics of the disease in a human-camel population within the KSA. Rigorous analysis of the model, which was fitted and cross-validated using the observed MERS-CoV data for the KSA, showed that its disease-free equilibrium was locally asymptotically stable whenever its reproduction number (denoted by $ {\mathbb R}_{0M} $) was less than unity. Using the fixed and estimated parameters of the model, the value of $ {\mathbb R}_{0M} $ for the KSA was estimated to be 0.84, suggesting that the prospects for MERS-CoV elimination are highly promising. The model was extended to allow for the assessment of public health intervention strategies, notably the potential use of vaccines for both humans and camels and the use of face masks by humans in public or when in close proximity with camels. Simulations of the extended model showed that the use of the face mask by humans who come in close proximity with camels, as a sole public health intervention strategy, significantly reduced human-to-camel and camel-to-human transmission of the disease, and this reduction depends on the efficacy and coverage of the mask type used in the community. For instance, if surgical masks are prioritized, the disease can be eliminated in both the human and camel population if at least 45% of individuals who have close contact with camels wear them consistently. The simulations further showed that while vaccinating humans as a sole intervention strategy only had marginal impact in reducing the disease burden in the human population, an intervention strategy based on vaccinating camels only resulted in a significant reduction in the disease burden in camels (and, consequently, in humans as well). Thus, this study suggests that attention should be focused on effectively combating the disease in the camel population, rather than in the human population. Furthermore, the extended model was used to simulate a hybrid strategy, which combined vaccination of both humans and camels as well as the use of face masks by humans. This simulation showed a marked reduction of the disease burden in both humans and camels, with an increasing effectiveness level of this intervention, in comparison to the baseline scenario or any of the aforementioned sole vaccination scenarios. In summary, this study showed that the prospect of the elimination of MERS-CoV-2 in the Kingdom of Saudi Arabia is promising using pharmaceutical (vaccination) and nonpharmaceutical (mask) intervention strategies, implemented in isolation or (preferably) in combination, that are focused on reducing the disease burden in the camel population.
Collapse
Affiliation(s)
- Adel Alatawi
- Department of Mathematics, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
- Biodiversity Genomics Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abba B Gumel
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
- Department of Mathematics and Applied Mathematics, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
25
|
Moon S, Lee KW, Park M, Moon J, Park SH, Kim S, Hwang J, Yoon JW, Jeon SM, Kim JS, Jeon YJ, Kweon DH. 3-Fucosyllactose-mediated modulation of immune response against virus infection. Int J Antimicrob Agents 2024; 64:107187. [PMID: 38697577 DOI: 10.1016/j.ijantimicag.2024.107187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Viral pathogens, particularly influenza and SARS-CoV-2, pose a significant global health challenge. Given the immunomodulatory properties of human milk oligosaccharides, in particular 2'-fucosyllactose and 3-fucosyllactose (3-FL), we investigated their dietary supplementation effects on antiviral responses in mouse models. This study revealed distinct immune modulations induced by 3-FL. RNA-sequencing data showed that 3-FL increased the expression of interferon receptors, such as Interferon Alpha and Beta Receptor (IFNAR) and Interferon Gamma Receptor (IFNGR), while simultaneously downregulating interferons and interferon-stimulated genes, an effect not observed with 2'-fucosyllactose supplementation. Such modulation enhanced antiviral responses in both cell culture and animal models while attenuating pre-emptive inflammatory responses. Nitric oxide concentrations in 3-FL-supplemented A549 cells and mouse lung tissues were elevated exclusively upon infection, reaching 5.8- and 1.9-fold increases over control groups, respectively. In addition, 3-FL promoted leukocyte infiltration into the site of infection upon viral challenge. 3-FL supplementation provided protective efficacy against lethal influenza challenge in mice. The demonstrated antiviral efficacy spanned multiple influenza strains and extended to SARS-CoV-2. In conclusion, 3-FL is a unique immunomodulator that helps protect the host from viral infection while suppressing inflammation prior to infection.
Collapse
Affiliation(s)
- Seokoh Moon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Myungseo Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeonghui Moon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Hee Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Soomin Kim
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jaehyeon Hwang
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jong-Won Yoon
- Advanced Protein Technologies Corp., Suwon, Republic of Korea
| | - Seon-Min Jeon
- Advanced Protein Technologies Corp., Suwon, Republic of Korea
| | - Jun-Seob Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea.
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea; Advanced Protein Technologies Corp., Suwon, Republic of Korea.
| |
Collapse
|
26
|
Krismer L, Schöppe H, Rauch S, Bante D, Sprenger B, Naschberger A, Costacurta F, Fürst A, Sauerwein A, Rupp B, Kaserer T, von Laer D, Heilmann E. Study of key residues in MERS-CoV and SARS-CoV-2 main proteases for resistance against clinically applied inhibitors nirmatrelvir and ensitrelvir. NPJ VIRUSES 2024; 2:23. [PMID: 38933182 PMCID: PMC11196219 DOI: 10.1038/s44298-024-00028-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/14/2024] [Indexed: 06/28/2024]
Abstract
The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an epidemic, zoonotically emerging pathogen initially reported in Saudi Arabia in 2012. MERS-CoV has the potential to mutate or recombine with other coronaviruses, thus acquiring the ability to efficiently spread among humans and become pandemic. Its high mortality rate of up to 35% and the absence of effective targeted therapies call for the development of antiviral drugs for this pathogen. Since the beginning of the SARS-CoV-2 pandemic, extensive research has focused on identifying protease inhibitors for the treatment of SARS-CoV-2. Our intention was therefore to assess whether these protease inhibitors are viable options for combating MERS-CoV. To that end, we used previously established protease assays to quantify inhibition of SARS-CoV-2, MERS-CoV and other main proteases. Nirmatrelvir inhibited several of these proteases, whereas ensitrelvir was less broadly active. To simulate nirmatrelvir's clinical use against MERS-CoV and subsequent resistance development, we applied a safe, surrogate virus-based system. Using the surrogate virus, we previously selected hallmark mutations of SARS-CoV-2-Mpro, such as T21I, M49L, S144A, E166A/K/V and L167F. In the current study, we selected a pool of MERS-CoV-Mpro mutants, characterized the resistance and modelled the steric effect of catalytic site mutants S142G, S142R, S147Y and A171S.
Collapse
Affiliation(s)
- Laura Krismer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - Helge Schöppe
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, 6020 Austria
| | - Stefanie Rauch
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - David Bante
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - Bernhard Sprenger
- Institute of Biochemistry, University of Innsbruck, CMBI – Center for Molecular Biosciences Innsbruck, Innsbruck, 6020 Austria
| | - Andreas Naschberger
- Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology KAUST, Thuwal, Saudi Arabia
| | | | - Anna Fürst
- Institute of Molecular Immunology, Technical University of Munich, Munich, 81675 Germany
| | - Anna Sauerwein
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - Bernhard Rupp
- Division of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, 6020 Austria
| | - Dorothee von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| | - Emmanuel Heilmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, 6020 Austria
| |
Collapse
|
27
|
Jiang Y, Zhang G, Li L, Chen J, Hao P, Gao Z, Hao J, Xu Z, Wang M, Li C, Jin N. A novel host restriction factor MRPS6 mediates the inhibition of PDCoV infection in HIEC-6 cells. Front Immunol 2024; 15:1381026. [PMID: 38919620 PMCID: PMC11196785 DOI: 10.3389/fimmu.2024.1381026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction Porcine deltacoronavirus (PDCoV) is a zoonotic pathogen with a global distribution, capable of infecting both pigs and humans. To mitigate the risk of cross-species transmission and potential outbreaks, it is crucial to characterize novel antiviral genes, particularly those from human hosts. Methods This research used HIEC-6 to investigate PDCoV infection. HIEC-6 cells were infected with PDCoV. Samples were collected 48 h postinfection for proteomic analysis. Results We discovered differential expression of MRPS6 gene at 48 h postinfection with PDCoV in HIEC-6 cells. The gene expression initially increased but then decreased. To further explore the role of MRPS6 in PDCoV infection, we conducted experiments involving the overexpression and knockdown of this gene in HIEC-6 and Caco2 cells, respectively. Our findings revealed that overexpression of MRPS6 significantly inhibited PDCoV infection in HIEC-6 cells, while knockdown of MRPS6 in Caco2 cells led to a significant increase of virus titer. Furthermore, we investigated the correlation between PDCoV infection and the expression of MRPS6. Subsequent investigations demonstrated that MRPS6 exerted an augmentative effect on the production of IFN-β through interferon pathway activation, consequently impeding the progression of PDCoV infection in cellular systems. In conclusion, this study utilized proteomic analysis to investigate the differential protein expression in PDCoV-infected HIEC-6 cells, providing evidence for the first time that the MRPS6 gene plays a restrictive role in PDCoV virus infection. Discussion Our findings initially provide the validation of MRPS6 as an upstream component of IFN-β pathway, in the promotion of IRF3, IRF7, STAT1, STAT2 and IFN-β production of HIEC-6 via dual-activation from interferon pathway.
Collapse
Affiliation(s)
- Yuhang Jiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guoqing Zhang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jing Chen
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zihan Gao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jiayi Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhiqiang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Maopeng Wang
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Chang Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
28
|
Norman FF, Gonzalez-Sanz M. Pulmonary infections in the returning traveler. Curr Opin Pulm Med 2024; 30:243-251. [PMID: 38323419 DOI: 10.1097/mcp.0000000000001051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
PURPOSE OF REVIEW The recent COVID-19 pandemic has shaped the epidemiology of other infectious diseases globally. International tourist arrivals are increasing and recovering to prepandemic levels. This review focuses on respiratory infections in travelers, highlighting the characteristics of the main imported viral, bacterial, fungal, and parasitic infections with pulmonary involvement. RECENT FINDINGS A recent systematic review estimated a prevalence of respiratory symptoms in travelers of around 35%, increasing to nearly 65% in the context of mass gatherings. Common viral and bacterial pathogens account for the majority of respiratory infections with an identified cause; however, recent data focus on the need for surveillance of emerging infections such as MERS-CoV, henipaviruses and multidrug resistant bacteria, which may be spread through travel. Fungal and parasitic respiratory infections are less common, and acquisition is usually associated with specific risk factors or exposure in endemic areas. Special risk groups, such as immunocompromised travelers, may be particularly vulnerable, presenting with severe disease or reactivation of latent infections. SUMMARY The next significant international epidemic could involve another new infectious agent causing respiratory disease and spreading via mobile populations. Official protocols should be adhered to, and public health interventions implemented for effective control. Continued and globally coordinated investments in research for new vaccines, therapeutic agents, disease modeling, and digital tracking strategies are essential.
Collapse
Affiliation(s)
- Francesca F Norman
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS
- Universidad de Alcalá
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Gonzalez-Sanz
- National Referral Unit for Tropical Diseases, Infectious Diseases Department, Ramón y Cajal University Hospital, IRYCIS
| |
Collapse
|
29
|
Gao F, Lin W, Wang X, Liao M, Zhang M, Qin N, Chen X, Xia L, Chen Q, Sha O. Identification of receptors and factors associated with human coronaviruses in the oral cavity using single-cell RNA sequencing. Heliyon 2024; 10:e28280. [PMID: 38560173 PMCID: PMC10981076 DOI: 10.1016/j.heliyon.2024.e28280] [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: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) ravaged the world, and Coronavirus Disease 2019 (COVID-19) exhibited highly prevalent oral symptoms that had significantly impacted the lives of affected patients. However, the involvement of four human coronavirus (HCoVs), namely SARS-CoV-2, SARS-CoV, MERS-CoV, and HCoV-229E, in oral cavity infections remained poorly understood. We integrated single-cell RNA sequencing (scRNA-seq) data of seven human oral tissues through consistent normalization procedure, including minor salivary gland (MSG), parotid gland (PG), tongue, gingiva, buccal, periodontium and pulp. The Seurat, scDblFinder, Harmony, SingleR, Ucell and scCancer packages were comprehensively used for analysis. We identified specific cell clusters and generated expression profiles of SARS-CoV-2 and coronavirus-associated receptors and factors (SCARFs) in seven oral regions, providing direction for predicting the tropism of four HCoVs for oral tissues, as well as for dental clinical treatment. Based on our analysis, it appears that various SCARFs, including ACE2, ASGR1, KREMEN1, DPP4, ANPEP, CD209, CLEC4G/M, TMPRSS family proteins (including TMPRSS2, TMPRSS4, and TMPRSS11A), and FURIN, are expressed at low levels in the oral cavity. Conversely, BSG, CTSB, and CTSL exhibit enrichment in oral tissues. Our study also demonstrates widespread expression of restriction factors, particularly IFITM1-3 and LY6E, in oral cells. Additionally, some replication, assembly, and trafficking factors appear to exhibit broad oral tissues expression patterns. Overall, the oral cavity could potentially serve as a high-risk site for SARS-CoV-2 infection, while displaying a comparatively lower degree of susceptibility towards other HCoVs (including SARS-CoV, MERS-CoV and HCoV-229E). Specifically, MSG, tongue, and gingiva represent potential sites of vulnerability for four HCoVs infection, with the MSG exhibiting a particularly high susceptibility. However, the expression patterns of SCARFs in other oral sites demonstrate relatively intricate and may only be specifically associated with SARS-CoV-2 infection. Our study sheds light on the mechanisms of HCoVs infection in the oral cavity as well as gains insight into the characteristics and distribution of possible HCoVs target cells in oral tissues, providing potential therapeutic targets for HCoVs infection in the oral cavity.
Collapse
Affiliation(s)
- Feng Gao
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| | - Weiming Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xia Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- The Chinese University of Hong Kong Shenzhen, School of Medicine, Shenzhen, China
| | - Mingfeng Liao
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Mingxia Zhang
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Nianhong Qin
- Department of Stomatology, Shenzhen People's Hospital, Shenzhen, China
| | - Xianxiong Chen
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lixin Xia
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Ou Sha
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| |
Collapse
|
30
|
Sayed AA. Evaluating COVID-19 vaccine acceptance among parents in Saudi Arabia: a systematic review examining attitudes, hesitancy, and intentions. Front Public Health 2024; 12:1327944. [PMID: 38584927 PMCID: PMC10995243 DOI: 10.3389/fpubh.2024.1327944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction The COVID-19 pandemic, affecting adults and children equally, has caused significant disruption to countries worldwide, including Saudi Arabia. In Saudi Arabia, the fast preventative measures and mass vaccine enrollment were vital to contain the devastating impact of the pandemic. However, vaccine hesitancy, especially among parents toward vaccinating their children, was a significant obstacle to vaccine uptake. Methods This systematic review followed PRISMA guidelines to assess parental willingness to vaccinate their children against COVID-19, determine the key determinants influencing such intention and attitudes, and underline the significant concerns and misconceptions regarding the vaccine among parents. The Joanne Briggs Institute (JBI) checklist for prevalence studies was used to assess included studies for risk of bias. Results Twenty-three studies were included in this systematic review, representing a total of 20,926 participants, with over 66% of them were female. Over 37% of the participants were willing to vaccinate their children against COVID-19. Parents' age, gender, level of education, and income were the main determinants of their intention to vaccinate their children. The parents' main concerns were the potential vaccine side effects, safety, and efficacy. Major misconceptions about the COVID-19 vaccine included it being dangerous to children and that children are at lower risk of severe infection; hence, vaccines were not needed. Discussion This seminal review provides insights to public health policymakers, which should be considered and taken together in light of other studies addressing parental vaccine hesitancy.
Collapse
Affiliation(s)
- Anwar A. Sayed
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
31
|
Abuyousef S, Alnaimi S, Omar NE, Elajez R, Elmekaty E, Abdelfattah-Arafa E, Barazi R, Ghasoub R, Rahhal A, Hamou F, Al-Amri M, Karawia A, Ajaj F, Alkhawaja R, Kardousha A, Awaisu A, Abou-Ali A, Khatib M, Aboukamar M, Al-Hail M. Early predictors of intensive care unit admission among COVID-19 patients in Qatar. Front Public Health 2024; 12:1278046. [PMID: 38572008 PMCID: PMC10987715 DOI: 10.3389/fpubh.2024.1278046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Background COVID-19 is associated with significant morbidity and mortality. This study aimed to explore the early predictors of intensive care unit (ICU) admission among patients with COVID-19. Methods This was a case-control study of adult patients with confirmed COVID-19. Cases were defined as patients admitted to ICU during the period February 29-May 29, 2020. For each case enrolled, one control was matched by age and gender. Results A total of 1,560 patients with confirmed COVID-19 were included. Each group included 780 patients with a predominant male gender (89.7%) and a median age of 49 years (interquartile range = 18). Predictors independently associated with ICU admission were cardiovascular disease (adjusted odds ratio (aOR) = 1.64, 95% confidence interval (CI): 1.16-2.32, p = 0.005), diabetes (aOR = 1.52, 95% CI: 1.08-2.13, p = 0.016), obesity (aOR = 1.46, 95% CI: 1.03-2.08, p = 0.034), lymphopenia (aOR = 2.69, 95% CI: 1.80-4.02, p < 0.001), high AST (aOR = 2.59, 95% CI: 1.53-4.36, p < 0.001), high ferritin (aOR = 1.96, 95% CI: 1.40-2.74, p < 0.001), high CRP (aOR = 4.09, 95% CI: 2.81-5.96, p < 0.001), and dyspnea (aOR = 2.50, 95% CI: 1.77-3.54, p < 0.001). Conclusion Having cardiovascular disease, diabetes, obesity, lymphopenia, dyspnea, and increased AST, ferritin, and CRP were independent predictors for ICU admission in patients with COVID-19.
Collapse
Affiliation(s)
- Safae Abuyousef
- Department of Pharmacy, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Shaikha Alnaimi
- Department of Pharmacy, Hamad Bin Khalifa Medical City, Hamad Medical Corporation, Doha, Qatar
| | - Nabil E. Omar
- Department of Pharmacy, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Health Sciences Program, Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| | - Reem Elajez
- Department of Pharmacy, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Eman Elmekaty
- Department of Pharmacy, Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | | | - Raja Barazi
- Department of Pharmacy, Al Wakra Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Rola Ghasoub
- Department of Pharmacy, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Ala Rahhal
- Department of Pharmacy, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Hamou
- Department of Pharmacy, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Maha Al-Amri
- Department of Pharmacy, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Karawia
- Department of Pharmacy, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Fatima Ajaj
- Department of Pharmacy, Home Health Care, Hamad Medical Corporation, Doha, Qatar
| | - Raja Alkhawaja
- Department of Pharmacy, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Kardousha
- Department of Pharmacy, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Awaisu
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Adel Abou-Ali
- Astellas Pharma Global Development, Inc., Northbrook, IL, United States
| | - Mohamad Khatib
- Department of Critical Care, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Mohammed Aboukamar
- Department of Infectious Disease, Communicable Diseases Center, Hamad Medical Corporation, Doha, Qatar
| | - Moza Al-Hail
- Department of Pharmacy, Women’s Wellness and Research Center, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
32
|
Xia D, Lu Z, Li S, Fang P, Yang C, He X, You Q, Sun G. Development of an Intelligent Reactive Oxygen Species-Responsive Dual-Drug Delivery Nanoplatform for Enhanced Precise Therapy of Acute Lung Injury. Int J Nanomedicine 2024; 19:2179-2197. [PMID: 38476280 PMCID: PMC10929269 DOI: 10.2147/ijn.s442727] [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: 10/04/2023] [Accepted: 02/06/2024] [Indexed: 03/14/2024] Open
Abstract
Introduction Acute lung injury (ALI) and its most severe form acute respiratory distress syndrome (ARDS) are commonly occurring devastating conditions that seriously threaten the respiratory system in critically ill patients. The current treatments improve oxygenation in patients with ALI/ARDS in the short term, but do not relieve the clinical mortality of patients with ARDS. Purpose To develop the novel drug delivery systems that can enhance the therapeutic efficacy of ALI/ARDS and impede adverse effects of drugs. Methods Based on the key pathophysiological process of ARDS that is the disruption of the pulmonary endothelial barrier, bilirubin (Br) and atorvastatin (As) were encapsulated into an intelligent reactive oxygen species (ROS)-responsive nanocarrier DSPE-TK-PEG (DPTP) to form nanoparticles (BA@DPTP) in which the thioketal bonds could be triggered by high ROS levels in the ALI tissues. Results BA@DPTP could accumulate in inflammatory pulmonary sites through passive targeting strategy and intelligently release Br and As only in the inflammatory tissue via ROS-responsive bond, thereby enhancing the drugs effectiveness and markedly reducing side effects. BA@DPTP effectively inhibited NF-κB signaling and NLRP3/caspase-1/GSDMD-dependent pyroptosis in mouse pulmonary microvascular endothelial cells. BA@DPTP not only protected mice with lipopolysaccharide-induced ALI and retained the integrity of the pulmonary structure, but also reduced ALI-related mortality. Conclusion This study combined existing drugs with nano-targeting strategies to develop a novel drug-targeting platform for the efficient treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Dunling Xia
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Zongqing Lu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Shuai Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Pu Fang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chun Yang
- Department of Emergency Intensive Care Unit, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei, People’s Republic of China
| | - Qinghai You
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| |
Collapse
|
33
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
34
|
Mallhi TH, Khan YH, Salman M, Butt MH. Are we neglecting MERS-CoV during COVID-19 pandemic: a need for refocusing on surveillance of the deadly MERS-CoV infection. Clin Microbiol Infect 2024; 30:256-257. [PMID: 37924970 DOI: 10.1016/j.cmi.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/07/2023] [Accepted: 10/21/2023] [Indexed: 11/06/2023]
Affiliation(s)
- Tauqeer Hussain Mallhi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia.
| | - Yusra Habib Khan
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia.
| | - Muhammad Salman
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore, Pakistan.
| | - Muhammad Hammad Butt
- Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
35
|
Abbas AT, El-Kafrawy SA, Tabll AA, Hashem AM, Al Subhi TL, Alsaadi M, Azhar EI. Development and characterization of three novel mouse monoclonal antibodies targeting spike protein S1 subunit of Middle East respiratory syndrome corona virus. Hum Antibodies 2024; 32:129-137. [PMID: 38758996 DOI: 10.3233/hab-240016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
BACKGROUND Middle East Respiratory Syndrome Coronavirus is a highly pathogenic virus that poses a significant threat to public health. OBJECTIVE The purpose of this study is to develop and characterize novel mouse monoclonal antibodies targeting the spike protein S1 subunit of the Middle East Respiratory Syndrome Corona Virus (MERS-CoV). METHODS In this study, three mouse monoclonal antibodies (mAbs) against MERS-CoV were generated and characterized using hybridoma technology. The mAbs were evaluated for their reactivity and neutralization activity. The mAbs were generated through hybridoma technology by the fusion of myeloma cells and spleen cells from MERS-CoV-S1 immunized mice. The resulting hybridomas were screened for antibody production using enzyme-linked immunosorbent assays (ELISA). RESULTS ELISA results demonstrated that all three mAbs exhibited strong reactivity against the MERS-CoV S1-antigen. Similarly, dot-ELISA revealed their ability to specifically recognize viral components, indicating their potential for diagnostic applications. Under non-denaturing conditions, Western blot showed the mAbs to have robust reactivity against a specific band at 116 KDa, corresponding to a putative MERS-CoV S1-antigen. However, no reactive bands were observed under denaturing conditions, suggesting that the antibodies recognize conformational epitopes. The neutralization assay showed no in vitro reactivity against MERS-CoV. CONCLUSION This study successfully generated three mouse monoclonal antibodies against MERS-CoV using hybridoma technology. The antibodies exhibited strong reactivity against MERS-CoV antigens using ELISA and dot ELISA assays. Taken together, these findings highlight the significance of these mAbs for potential use as valuable tools for MERS-CoV research and diagnosis (community and field-based surveillance and viral antigen detection).
Collapse
Affiliation(s)
- Aymn T Abbas
- Special Infectious Agents Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif A El-Kafrawy
- Special Infectious Agents Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashraf A Tabll
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
- Egypt Centre for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tagreed L Al Subhi
- Special Infectious Agents Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Alsaadi
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Xiao S, Yuan Z, Huang Y. The Potential Role of Nitric Oxide as a Therapeutic Agent against SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:17162. [PMID: 38138990 PMCID: PMC10742813 DOI: 10.3390/ijms242417162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the greatest worldwide public health threat of this century, which may predispose multi-organ failure (especially the lung) and death despite numerous mild and moderate symptoms. Recent studies have unraveled the molecular and clinical characteristics of the infectivity, pathogenicity, and immune evasion of SARS-CoV-2 and thus improved the development of many different therapeutic strategies to combat COVID-19, including treatment and prevention. Previous studies have indicated that nitric oxide (NO) is an antimicrobial and anti-inflammatory molecule with key roles in pulmonary vascular function in the context of viral infections and other pulmonary disease states. This review summarized the recent advances of the pathogenesis of SARS-CoV-2, and accordingly elaborated on the potential application of NO in the management of patients with COVID-19 through antiviral activities and anti-inflammatory properties, which mitigate the propagation of this disease. Although there are some limits of NO in the treatment of COVID-19, it might be a worthy candidate in the multiple stages of COVID-19 prevention or therapy.
Collapse
Affiliation(s)
| | | | - Yi Huang
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan 430020, China
| |
Collapse
|
37
|
Dong J, Wu X, Hu Q, Sun C, Li J, Song P, Su Y, Zhou L. An immobilization-free electrochemical biosensor based on CRISPR/Cas13a and FAM-RNA-MB for simultaneous detection of multiple pathogens. Biosens Bioelectron 2023; 241:115673. [PMID: 37717422 DOI: 10.1016/j.bios.2023.115673] [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: 06/21/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
To better respond to biosecurity issues, we need to build good technology and material reserves for pathogenic microorganism screening. Here, we designed an electrochemical/optical signal probe with a common fluorophore and an electrochemically active group, breaking the previous perception that the signal probe is composed of a fluorophore and a quenching group and realizing the response of three signals: electrochemistry, fluorescence, and direct observation. Then, we proposed a homogeneous electrochemical nucleic acid detection system based on CRISPR/Cas named "HELEN-CR" by integrating free electrochemical/optical signal probes and Cas13a cleavage, achieving a limit of detection of 1 pM within 25 min. To improve the detection sensitivity, we applied recombinase polymerase amplification to amplify the target nucleic acid, achieving a limit of detection of 30 zM within 45 min. Complemented by our self-developed multi-chamber microfluidic chip and portable electrochemical instrument, simultaneous detection of multiple pathogens can be achieved within 50 min, facilitating minimally trained personnel to obtain detection results quickly in a difficult environment. This study proposes a simple, scalable, and general idea and solution for the rapid detection of pathogenic microorganisms and biosecurity monitoring.
Collapse
Affiliation(s)
- Jinying Dong
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoya Wu
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiushi Hu
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China; Biosafety Research Center Yangtze River Delta in Zhangjiagang, Suzhou, 215611, China
| | - Chongsi Sun
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiahao Li
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China
| | - Peng Song
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yan Su
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lei Zhou
- National Key Laboratory of Biochemical Engineering, PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190, China; Biosafety Research Center Yangtze River Delta in Zhangjiagang, Suzhou, 215611, China.
| |
Collapse
|
38
|
Saksena NK, Reddy SB, Miranda-Saksena M, Cardoso THS, Silva EMA, Ferreira JC, Rabeh WM. SARS-CoV-2 variants, its recombinants and epigenomic exploitation of host defenses. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166836. [PMID: 37549720 DOI: 10.1016/j.bbadis.2023.166836] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
Since 2003, we have seen the emergence of novel viruses, such as SARS-CoV-1, MERS, ZIKA, swine flu virus H1N1, Marburg, Monkeypox, Ebola, and SARS-CoV-2, but none of them gained pandemic proportions similar to SARS-CoV-2. This could be attributed to unique viral traits, allowing its rapid global dissemination following its emergence in October 2019 in Wuhan, China, which appears to be primarily driven by the emergence of highly transmissible and virulent variants that also associate, in some cases, with severe disease and considerable mortality caused by fatal pneumonia, acute respiratory distress syndrome (ARDS) in infected individuals. Mechanistically, several factors are involved in viral pathogenesis, and epigenetic alterations take the front seat in host-virus interactions. The molecular basis of all viral infections, including SARS-CoV-2, tightly hinges on the transitory silencing of the host gene machinery via epigenetic modulation. SARS-CoV-2 also hijacks and subdues the host gene machinery, leading to epigenetic modulation of the critical host elements responsible for antiviral immunity. Epigenomics is a powerful, unexplored avenue that can provide a profound understanding of virus-host interactions and lead to the development of epigenome-based therapies and vaccines to counter viruses. This review discusses current developments in SARS-CoV-2 variation and its role in epigenetic modulation in infected hosts. This review provides an overview, especially in the context of emerging viral strains, their recombinants, and their possible roles in the epigenetic exploitation of host defense and viral pathogenesis. It provides insights into host-virus interactions at the molecular, genomic, and immunological levels and sheds light on the future of epigenomics-based therapies for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nitin K Saksena
- Victoria University, Footscray Campus, Melbourne, VIC. Australia.
| | - Srinivasa Bonam Reddy
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Thyago H S Cardoso
- OMICS Centre of Excellence, G42 Healthcare, Mazdar City, Abu Dhabi, United Arab Emirates.
| | - Edson M A Silva
- Science Division, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana C Ferreira
- Science Division, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates.
| | - Wael M Rabeh
- Science Division, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
39
|
Li X, Song Y. Structure and function of SARS-CoV and SARS-CoV-2 main proteases and their inhibition: A comprehensive review. Eur J Med Chem 2023; 260:115772. [PMID: 37659195 PMCID: PMC10529944 DOI: 10.1016/j.ejmech.2023.115772] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023]
Abstract
Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) identified in 2003 infected ∼8000 people in 26 countries with 800 deaths, which was soon contained and eradicated by syndromic surveillance and enhanced quarantine. A closely related coronavirus SARS-CoV-2, the causative agent of COVID-19 identified in 2019, has been dramatically more contagious and catastrophic. It has infected and caused various flu-like symptoms of billions of people in >200 countries, including >6 million people died of or with the virus. Despite the availability of several vaccines and antiviral drugs against SARS-CoV-2, finding new therapeutics is needed because of viral evolution and a possible emerging coronavirus in the future. The main protease (Mpro) of these coronaviruses plays important roles in their life cycle and is essential for the viral replication. This article represents a comprehensive review of the function, structure and inhibition of SARS-CoV and -CoV-2 Mpro, including structure-activity relationships, protein-inhibitor interactions and clinical trial status.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - Yongcheng Song
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
40
|
de Almeida MT, Barbosa AP, Bomfim CG, Visnardi AB, Vinces TC, Ceroni A, Durigon EL, Guzzo CR. Obtaining a high titer of polyclonal antibodies from rats to the SARS-CoV-2 nucleocapsid protein and its N- and C-terminal domains for diagnostic test development. J Immunol Methods 2023; 522:113558. [PMID: 37704125 DOI: 10.1016/j.jim.2023.113558] [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: 04/18/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped, plus-stranded RNA virus responsible for the Coronavirus Disease 2019 (COVID-19). Patients infected with COVID-19 may be asymptomatic or have symptoms ranging from mild manifestations to severe cases of the disease that could lead to death. The SARS-CoV-2 genome encodes 4 structural proteins, including the Spike protein (S), the Nucleocapsid protein (N), Membrane protein (M) and, the Envelope protein (E). The N protein forms a major component of the ribonucleoprotein complex within the virus particle and play a vital role in its transcription and replication. Nevertheless, the S protein was the most important protein in the development of vaccines against COVID-19. However, the decrease in number of registered immunizations against the disease and the rapid drop in neutralizing antibody titers together with looser preventive measures for virus transmission, favored the rapid appearance of new variants of concerns (VOCs) that primarily show mutations in the S protein. This fact makes the N protein a good candidate for the development of diagnostic tests, due to its stability, amino acid conservation, high immunogenicity, and the smaller likelihood of mutation. With the aim of developing a new diagnostic kit based on the N protein, we evaluated the humoral response in female Wistar rats against this target. Three constructions of the N protein were used to inoculate the animals: the full-length protein (Cfull), the N- (NTD), and the C-terminal (CTD) portion of the protein. The immunizations induced the animal's immune response, with specific polyclonal IgG antibodies against the Cfull protein and its fragments. There were not non-specific bind to the protein used as negative control. Anti-Cfull antibodies demonstrated high efficiency in binding to the NTD protein and the antibodies present in the anti-CTD and anti-NTD sera have recognized the Cfull protein, but they were not able to recognize the NTD and CTD proteins, respectively. Our results indicate an efficient protocol for obtaining high antibody titers against the N recombinant protein of SARS-CoV-2 and its fragments highlighting the Cfull protein, which can be used in the development of new diagnostic kits.
Collapse
Affiliation(s)
| | - Ana Paula Barbosa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila Gasque Bomfim
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline Biazola Visnardi
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tania Churasacari Vinces
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexandre Ceroni
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edison Luiz Durigon
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur USP, São Paulo, Brazil
| | - Cristiane Rodrigues Guzzo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
41
|
Martinez DR, Schäfer A, Gavitt TD, Mallory ML, Lee E, Catanzaro NJ, Chen H, Gully K, Scobey T, Korategere P, Brown A, Smith L, Parks R, Barr M, Newman A, Bowman C, Powers JM, Soderblom EJ, Mansouri K, Edwards RJ, Baric RS, Haynes BF, Saunders KO. Vaccine-mediated protection against Merbecovirus and Sarbecovirus challenge in mice. Cell Rep 2023; 42:113248. [PMID: 37858337 PMCID: PMC10842144 DOI: 10.1016/j.celrep.2023.113248] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/30/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
The emergence of three highly pathogenic human coronaviruses-severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003, Middle Eastern respiratory syndrome (MERS)-CoV in 2012, and SARS-CoV-2 in 2019-underlines the need to develop broadly active vaccines against the Merbecovirus and Sarbecovirus betacoronavirus subgenera. While SARS-CoV-2 vaccines protect against severe COVID-19, they do not protect against other sarbecoviruses or merbecoviruses. Here, we vaccinate mice with a trivalent sortase-conjugate nanoparticle (scNP) vaccine containing the SARS-CoV-2, RsSHC014, and MERS-CoV receptor-binding domains (RBDs), which elicited live-virus neutralizing antibody responses. The trivalent RBD scNP elicited serum neutralizing antibodies against bat zoonotic Wuhan Institute of Virology-1 (WIV-1)-CoV, SARS-CoV, SARS-CoV-2 BA.1, SARS-CoV-2 XBB.1.5, and MERS-CoV live viruses. The monovalent SARS-CoV-2 RBD scNP vaccine only protected against Sarbecovirus challenge, whereas the trivalent RBD scNP vaccine protected against both Merbecovirus and Sarbecovirus challenge in highly pathogenic and lethal mouse models. This study demonstrates proof of concept for a single pan-sarbecovirus/pan-merbecovirus vaccine that protects against three highly pathogenic human coronaviruses spanning two betacoronavirus subgenera.
Collapse
Affiliation(s)
- David R Martinez
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA; Yale Center for Infection and Immunity, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tyler D Gavitt
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Esther Lee
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nicholas J Catanzaro
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haiyan Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kendra Gully
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Trevor Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pooja Korategere
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Alecia Brown
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lena Smith
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amanda Newman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Cindy Bowman
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erik J Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC 27710, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
42
|
Soni MK, Migliori E, Fu J, Assal A, Chan HT, Pan J, Khatiwada P, Ciubotariu R, May MS, Pereira MR, De Giorgi V, Sykes M, Mapara MY, Muranski PJ. The prospect of universal coronavirus immunity: characterization of reciprocal and non-reciprocal T cell responses against SARS-CoV2 and common human coronaviruses. Front Immunol 2023; 14:1212203. [PMID: 37901229 PMCID: PMC10612330 DOI: 10.3389/fimmu.2023.1212203] [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: 04/25/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
T cell immunity plays a central role in clinical outcomes of Coronavirus Infectious Disease 2019 (COVID-19) and T cell-focused vaccination or cellular immunotherapy might provide enhanced protection for some immunocompromised patients. Pre-existing T cell memory recognizing SARS-CoV-2 antigens antedating COVID-19 infection or vaccination, may have developed as an imprint of prior infections with endemic non-SARS human coronaviruses (hCoVs) OC43, HKU1, 229E, NL63, pathogens of "common cold". In turn, SARS-CoV-2-primed T cells may recognize emerging variants or other hCoV viruses and modulate the course of subsequent hCoV infections. Cross-immunity between hCoVs and SARS-CoV-2 has not been well characterized. Here, we systematically investigated T cell responses against the immunodominant SARS-CoV-2 spike, nucleocapsid and membrane proteins and corresponding antigens from α- and β-hCoVs among vaccinated, convalescent, and unexposed subjects. Broad T cell immunity against all tested SARS-CoV-2 antigens emerged in COVID-19 survivors. In convalescent and in vaccinated individuals, SARS-CoV-2 spike-specific T cells reliably recognized most SARS-CoV-2 variants, however cross-reactivity against the omicron variant was reduced by approximately 47%. Responses against spike, nucleocapsid and membrane antigens from endemic hCoVs were significantly more extensive in COVID-19 survivors than in unexposed subjects and displayed cross-reactivity between α- and β-hCoVs. In some, non-SARS hCoV-specific T cells demonstrated a prominent non-reciprocal cross-reactivity with SARS-CoV-2 antigens, whereas a distinct anti-SARS-CoV-2 immunological repertoire emerged post-COVID-19, with relatively limited cross-recognition of non-SARS hCoVs. Based on this cross-reactivity pattern, we established a strategy for in-vitro expansion of universal anti-hCoV T cells for adoptive immunotherapy. Overall, these results have implications for the future design of universal vaccines and cell-based immune therapies against SARS- and non-SARS-CoVs.
Collapse
Affiliation(s)
- Mithil K. Soni
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Edoardo Migliori
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Amer Assal
- Department of Medicine, Blood and Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY, United States
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Hei Ton Chan
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Jian Pan
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Prabesh Khatiwada
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Rodica Ciubotariu
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Michael S. May
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Marcus R. Pereira
- Department of Medicine, Division of Infectious Disease, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Valeria De Giorgi
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Markus Y. Mapara
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Pawel J. Muranski
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
43
|
Zhan D, Li D, Yuan K, Sun Y, He L, Zhong J, Wang L. Characteristics of the pulmonary microbiota in patients with mild and severe pulmonary infection. Front Cell Infect Microbiol 2023; 13:1227581. [PMID: 37900322 PMCID: PMC10602873 DOI: 10.3389/fcimb.2023.1227581] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023] Open
Abstract
Background Lung infection is a global health problem associated with high morbidity and mortality and increasing rates of hospitalization. The correlation between pulmonary microecology and infection severity remains unclear. Therefore, the purpose of this study was to investigate the differences in lung microecology and potential biomarkers in patients with mild and severe pulmonary infection. Method Patients with pulmonary infection or suspected infection were divided into the mild group (140 cases) and the severe group (80 cases) according to pneomonia severity index (PSI) scores. Here, we used metagenomic next-generation sequencing (mNGS) to detect DNA mainly from bronchoalveolar lavage fluid (BALF) collected from patients to analyze changes in the lung microbiome of patients with different disease severity. Result We used the mNGS to analyze the pulmonary microecological composition in patients with pulmonary infection. The results of alpha diversity and beta diversity analysis showed that the microbial composition between mild and severe groups was similar on the whole. The dominant bacteria were Acinetobacter, Bacillus, Mycobacterium, Staphylococcus, and Prevotella, among others. Linear discriminant analysis effect size (LEfSe) results showed that there were significant differences in virus composition between the mild and severe patients, especially Simplexvirus and Cytomegalovirus, which were prominent in the severe group. The random forest model screened 14 kinds of pulmonary infection-related pathogens including Corynebacterium, Mycobacterium, Streptococcus, Klebsiella, and Acinetobacter. In addition, it was found that Rothia was negatively correlated with Acinetobacter, Mycobacterium, Bacillus, Enterococcus, and Klebsiella in the mild group through co-occurrence network, while no significant correlation was found in the severe group. Conclusion Here, we describe the composition and diversity of the pulmonary microbiome in patients with pulmonary infection. A significant increase in viral replication was found in the severe group, as well as a significant difference in microbial interactions between patients with mild and severe lung infections, particularly the association between the common pathogenic bacteria and Rothia. This suggests that both pathogen co-viral infection and microbial interactions may influence the course of disease. Of course, more research is needed to further explore the specific mechanisms by which microbial interactions influence disease severity.
Collapse
Affiliation(s)
- Danting Zhan
- Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Guangdong, China
| | - Dan Li
- BGI Genomics, Shenzhen, China
| | - Ke Yuan
- BGI Genomics, Shenzhen, China
| | | | | | - Jiacheng Zhong
- Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Guangdong, China
| | - Lingwei Wang
- Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Guangdong, China
| |
Collapse
|
44
|
Eain MMG, Nolan K, Murphy B, McCaul C, MacLoughlin R. Exhaled patient derived aerosol dispersion during awake tracheal intubation with concurrent high flow nasal therapy. J Clin Monit Comput 2023; 37:1265-1273. [PMID: 36930390 PMCID: PMC10022553 DOI: 10.1007/s10877-023-00990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023]
Abstract
Awake Tracheal Intubation (ATI) can be performed in cases where there is potential for difficult airway management. It is considered an aerosol generating procedure and is a source of concern to healthcare workers due to the risk of transmission of airborne viral infections, such as SARS-CoV-2. At present, there is a lack of data on the quantities, size distributions and spread of aerosol particles generated during such procedures. This was a volunteer observational study which took place in an operating room of a university teaching hospital. Optical particle sizers were used to provide real time aerosol characterisation during a simulated ATI performed with concurrent high-flow nasal oxygen therapy. The particle sizers were positioned at locations that represented the different locations of clinical staff in an operating room during an ATI. The greatest concentration of patient derived aerosol particles was within 0.5-1.0 m of the subject and along their midline, 2242 #/cm3. As the distance, both radial and longitudinal, from the subject increased, the concentration decreased towards ambient levels, 36.9 ± 5.1 #/cm3. Patient derived aerosol particles < 5 µm in diameter remained entrained in the exhaled aerosol plume and fell to the floor or onto the subject. Patient derived particles > 5 µm in diameter broke away from the exhaled plume and spread radially throughout the operating room. Irrespective of distance and ventilation status, full airborne protective equipment should be worn by all staff when ATI is being performed on patients with suspected viral respiratory infections.
Collapse
Affiliation(s)
- Marc Mac Giolla Eain
- Research and Development, Science and Emerging Technologies, Aerogen Ltd, IDA Business Park, Dangan, Galway, H91HE94, Ireland
| | - Kevin Nolan
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
| | - Brian Murphy
- Department of Anaesthesia, Rotunda Hospital, Parnell Square, Dublin, Ireland
| | - Conan McCaul
- Department of Anaesthesia, Rotunda Hospital, Parnell Square, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Ltd, IDA Business Park, Dangan, Galway, H91HE94, Ireland.
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland.
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland.
| |
Collapse
|
45
|
Tse LV, Hou YJ, McFadden E, Lee RE, Scobey TD, Leist SR, Martinez DR, Meganck RM, Schäfer A, Yount BL, Mascenik T, Powers JM, Randell SH, Zhang Y, Wang L, Mascola J, McLellan JS, Baric RS. A MERS-CoV antibody neutralizes a pre-emerging group 2c bat coronavirus. Sci Transl Med 2023; 15:eadg5567. [PMID: 37756379 PMCID: PMC11292784 DOI: 10.1126/scitranslmed.adg5567] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The repeated emergence of zoonotic human betacoronaviruses (β-CoVs) dictates the need for broad therapeutics and conserved epitope targets for countermeasure design. Middle East respiratory syndrome (MERS)-related coronaviruses (CoVs) remain a pressing concern for global health preparedness. Using metagenomic sequence data and CoV reverse genetics, we recovered a full-length wild-type MERS-like BtCoV/li/GD/2014-422 (BtCoV-422) recombinant virus, as well as two reporter viruses, and evaluated their human emergence potential and susceptibility to currently available countermeasures. Similar to MERS-CoV, BtCoV-422 efficiently used human and other mammalian dipeptidyl peptidase protein 4 (DPP4) proteins as entry receptors and an alternative DPP4-independent infection route in the presence of exogenous proteases. BtCoV-422 also replicated efficiently in primary human airway, lung endothelial, and fibroblast cells, although less efficiently than MERS-CoV. However, BtCoV-422 shows minor signs of infection in 288/330 human DPP4 transgenic mice. Several broad CoV antivirals, including nucleoside analogs and 3C-like/Mpro protease inhibitors, demonstrated potent inhibition against BtCoV-422 in vitro. Serum from mice that received a MERS-CoV mRNA vaccine showed reduced neutralizing activity against BtCoV-422. Although most MERS-CoV-neutralizing monoclonal antibodies (mAbs) had limited activity, one anti-MERS receptor binding domain mAb, JC57-11, neutralized BtCoV-422 potently. A cryo-electron microscopy structure of JC57-11 in complex with BtCoV-422 spike protein revealed the mechanism of cross-neutralization involving occlusion of the DPP4 binding site, highlighting its potential as a broadly neutralizing mAb for group 2c CoVs that use DPP4 as a receptor. These studies provide critical insights into MERS-like CoVs and provide candidates for countermeasure development.
Collapse
Affiliation(s)
- Longping V. Tse
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63014
| | - Yixuan J. Hou
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Elizabeth McFadden
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Rhianna E Lee
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Trevor D. Scobey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Rita M. Meganck
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO 63014
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Boyd L. Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Teresa Mascenik
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - John M. Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Yi Zhang
- National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, MD 20892
| | - Lingshu Wang
- National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, MD 20892
| | - John Mascola
- National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, MD 20892
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
46
|
Wang H, Xu J, Xiang L. Microneedle-Mediated Transcutaneous Immunization: Potential in Nucleic Acid Vaccination. Adv Healthc Mater 2023; 12:e2300339. [PMID: 37115817 DOI: 10.1002/adhm.202300339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Efforts aimed at exploring economical and efficient vaccination have taken center stage to combat frequent epidemics worldwide. Various vaccines have been developed for infectious diseases, among which nucleic acid vaccines have attracted much attention from researchers due to their design flexibility and wide application. However, the lack of an efficient delivery system considerably limits the clinical translation of nucleic acid vaccines. As mass vaccinations via syringes are limited by low patient compliance and high costs, microneedles (MNs), which can achieve painless, cost-effective, and efficient drug delivery, can provide an ideal vaccination strategy. The MNs can break through the stratum corneum barrier in the skin and deliver vaccines to the immune cell-rich epidermis and dermis. In addition, the feasibility of MN-mediated vaccination is demonstrated in both preclinical and clinical studies and has tremendous potential for the delivery of nucleic acid vaccines. In this work, the current status of research on MN vaccines is reviewed. Moreover, the improvements of MN-mediated nucleic acid vaccination are summarized and the challenges of its clinical translation in the future are discussed.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
47
|
François S, Nazki S, Vickers SH, Fournié G, Perrins CM, Broadbent AJ, Pybus OG, Hill SC. Genetic diversity, recombination and cross-species transmission of a waterbird gammacoronavirus in the wild. J Gen Virol 2023; 104. [PMID: 37589541 DOI: 10.1099/jgv.0.001883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Viruses emerging from wildlife can cause outbreaks in humans and domesticated animals. Predicting the emergence of future pathogens and mitigating their impacts requires an understanding of what shapes virus diversity and dynamics in wildlife reservoirs. In order to better understand coronavirus ecology in wild species, we sampled birds within a coastal freshwater lagoon habitat across 5 years, focussing on a large population of mute swans (Cygnus olor) and the diverse species that they interact with. We discovered and characterised the full genome of a divergent gammacoronavirus belonging to the Goose coronavirus CB17 species. We investigated the genetic diversity and dynamics of this gammacoronavirus using untargeted metagenomic sequencing of 223 faecal samples from swans of known age and sex, and RT-PCR screening of 1632 additional bird samples. The virus circulated persistently within the bird community; virus prevalence in mute swans exhibited seasonal variations, but did not change with swan age-class or epidemiological year. One whole genome was fully characterised, and revealed that the virus originated from a recombination event involving an undescribed gammacoronavirus species. Multiple lineages of this gammacoronavirus co-circulated within our study population. Viruses from this species have recently been detected in aquatic birds from both the Anatidae and Rallidae families, implying that host species habitat sharing may be important in shaping virus host range. As the host range of the Goose coronavirus CB17 species is not limited to geese, we propose that this species name should be updated to 'Waterbird gammacoronavirus 1'. Non-invasive sampling of bird coronaviruses may provide a tractable model system for understanding the evolutionary and cross-species dynamics of coronaviruses.
Collapse
Affiliation(s)
- Sarah François
- Department of Biology, University of Oxford, South Park Road, Oxford, OX1 3SY, UK
| | - Salik Nazki
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FZ, UK
- The Pirbright Institute, Ash Rd, Pirbright, Woking GU24 0NF, UK
| | - Stephen H Vickers
- Department of Pathobiology and Population Science, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| | - Guillaume Fournié
- Department of Pathobiology and Population Science, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
- Université de Lyon, INRAE, VetAgro Sup, UMR EPIA, Marcy l'Etoile, France
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMR EPIA, Saint-Gènes-Champanelle, France
| | | | - Andrew J Broadbent
- The Pirbright Institute, Ash Rd, Pirbright, Woking GU24 0NF, UK
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD20742, USA
| | - Oliver G Pybus
- Department of Biology, University of Oxford, South Park Road, Oxford, OX1 3SY, UK
- Department of Pathobiology and Population Science, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| | - Sarah C Hill
- Department of Pathobiology and Population Science, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| |
Collapse
|
48
|
Hemnani M, da Silva PG, Thompson G, Poeta P, Rebelo H, Mesquita JR. First Report of Alphacoronavirus Circulating in Cavernicolous Bats from Portugal. Viruses 2023; 15:1521. [PMID: 37515207 PMCID: PMC10384150 DOI: 10.3390/v15071521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The emergence of novel coronaviruses (CoVs) has emphasized the need to understand their diversity and distribution in animal populations. Bats have been identified as crucial reservoirs for CoVs, and they are found in various bat species worldwide. In this study, we investigated the presence of CoVs of four cavernicolous bats in six locations in the centre and south of Portugal. We collected faeces, anal, and buccal swab samples, as well as air samples from the locations using a Coriolis air sampler. Our results indicate that CoVs were more readily detected in faecal samples compared to anal and buccal swab samples. No CoVs were detected in the air samples. Phylogenetic analysis showed that the detected viruses belong to the Alphacoronavirus genus. This study represents the first report of Alphacoronaviruses circulating in bats in Portugal and highlights the importance of continuous surveillance for novel CoVs in bat populations globally. Ongoing surveillance for CoVs in bat populations is essential as they are a vital source of these viruses. It is crucial to understand the ecological relationships between animals, humans, and the environment to prevent and control the emergence and transmission of infectious diseases. Further ecological studies are needed to investigate the factors contributing to the emergence and transmission of zoonotic viruses.
Collapse
Affiliation(s)
- Mahima Hemnani
- School of Medicine and Biomedical Sciences, Porto University, 4050-313 Porto, Portugal; (M.H.); (P.G.d.S.); (G.T.)
| | - Priscilla Gomes da Silva
- School of Medicine and Biomedical Sciences, Porto University, 4050-313 Porto, Portugal; (M.H.); (P.G.d.S.); (G.T.)
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, 4099-002 Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), 4050-313 Porto, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnotlogy and Energy, Faculty of Engineering, University of Porto, 4099-002 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4099-002 Porto, Portugal
| | - Gertrude Thompson
- School of Medicine and Biomedical Sciences, Porto University, 4050-313 Porto, Portugal; (M.H.); (P.G.d.S.); (G.T.)
- Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Universidade do Porto, 4485-661 Vairão, Portugal;
| | - Patricia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os Montes e Alto Douro, 5000-801 Vila Real, Portugal;
- Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisbon, 1099-085 Caparica, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Veterinary and Animal Research Centre, Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000-801 Vila Real, Portugal
| | - Hugo Rebelo
- Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO Laboratório Associado, Universidade do Porto, 4485-661 Vairão, Portugal;
- ESS, Instituto Politécnico de Setúbal, 2910-761 Setúbal, Portugal
| | - João R. Mesquita
- School of Medicine and Biomedical Sciences, Porto University, 4050-313 Porto, Portugal; (M.H.); (P.G.d.S.); (G.T.)
- Epidemiology Research Unit (EPIunit), Institute of Public Health, University of Porto, 4099-002 Porto, Portugal
| |
Collapse
|
49
|
Ivachtchenko AV, Ivashchenko AA, Shkil DO, Ivashchenko IA. Aprotinin-Drug against Respiratory Diseases. Int J Mol Sci 2023; 24:11173. [PMID: 37446350 PMCID: PMC10342444 DOI: 10.3390/ijms241311173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Aprotinin (APR) was discovered in 1930. APR is an effective pan-protease inhibitor, a typical "magic shotgun". Until 2007, APR was widely used as an antithrombotic and anti-inflammatory drug in cardiac and noncardiac surgeries for reduction of bleeding and thus limiting the need for blood transfusion. The ability of APR to inhibit proteolytic activation of some viruses leads to its use as an antiviral drug for the prevention and treatment of acute respiratory virus infections. However, due to incompetent interpretation of several clinical trials followed by incredible controversy in the literature, the usage of APR was nearly stopped for a decade worldwide. In 2015-2020, after re-analysis of these clinical trials' data the restrictions in APR usage were lifted worldwide. This review discusses antiviral mechanisms of APR action and summarizes current knowledge and prospective regarding the use of APR treatment for diseases caused by RNA-containing viruses, including influenza and SARS-CoV-2 viruses, or as a part of combination antiviral treatment.
Collapse
Affiliation(s)
- Alexandre V. Ivachtchenko
- ChemDiv Inc., San Diego, CA 92130, USA; (A.A.I.); (I.A.I.)
- ASAVI LLC, 1835 East Hallandale Blvd #442, Hallandale Beach, FL 33009, USA;
| | | | - Dmitrii O. Shkil
- ASAVI LLC, 1835 East Hallandale Blvd #442, Hallandale Beach, FL 33009, USA;
| | | |
Collapse
|
50
|
Jagirdhar GSK, Pulakurthi YS, Chigurupati HD, Surani S. Gastrointestinal tract and viral pathogens. World J Virol 2023; 12:136-150. [PMID: 37396706 PMCID: PMC10311582 DOI: 10.5501/wjv.v12.i3.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/17/2023] [Accepted: 04/27/2023] [Indexed: 06/21/2023] Open
Abstract
Viral gastroenteritis is the most common viral illness that affects the gastrointestinal (GI) tract, causing inflammation and irritation of the lining of the stomach and intestines. Common signs and symptoms associated with this condition include abdominal pain, diarrhea, and dehydration. The infections commonly involved in viral gastroenteritis are rotavirus, norovirus, and adenovirus, which spread through the fecal-oral and contact routes and cause non-bloody diarrhea. These infections can affect both immunocompetent and immunocompromised individuals. Since the pandemic in 2019, coronavirus gastroenteritis has increased in incidence and prevalence. Morbidity and mortality rates from viral gastroenteritis have declined significantly over the years due to early recognition, treatment with oral rehydration salts, and prompt vaccination. Improved sanitation measures have also played a key role in reducing the transmission of infection. In addition to viral hepatitis causing liver disease, herpes virus, and cytomegalovirus are responsible for ulcerative GI disease. They are associated with bloody diarrhea and commonly occur in im-munocompromised individuals. Hepatitis viruses, Epstein-Barr virus, herpesvirus 8, and human papillomavirus have been involved in benign and malignant diseases. This mini review aims to list different viruses affecting the GI tract. It will cover common symptoms aiding in diagnosis and various important aspects of each viral infection that can aid diagnosis and management. This will help primary care physicians and hospitalists diagnose and treat patients more easily.
Collapse
Affiliation(s)
| | | | | | - Salim Surani
- Department of Pulmonary, Critical Care and Sleep Medicine, Texas A&M University, College Station, TX 77843, United States
| |
Collapse
|