1
|
Ulhe A, Raina P, Chaudhary A, Kaul-Ghanekar R. Alpha-linolenic acid-mediated epigenetic reprogramming of cervical cancer cell lines. Epigenetics 2025; 20:2451551. [PMID: 39895102 PMCID: PMC11792827 DOI: 10.1080/15592294.2025.2451551] [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: 05/30/2024] [Revised: 11/25/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Cervical cancer, the fourth most common cancer globally and the second most prevalent cancer among women in India, is primarily caused by Human Papilloma Virus (HPV). The association of diet with cancer etiology and prevention has been well established and nutrition has been shown to regulate cancer through modulation of epigenetic markers. Dietary fatty acids, especially omega-3, reduce the risk of cancer by preventing or reversing the progression through a variety of cellular targets, including epigenetic regulation. In this work, we have evaluated the potential of ALA (α linolenic acid), an ω-3 fatty acid, to regulate cervical cancer through epigenetic mechanisms. The effect of ALA was evaluated on the regulation of histone deacetylases1, DNA methyltransferases 1, and 3b, and global DNA methylation by ELISA. RT-PCR was utilized to assess the expression of tumor regulatory genes (hTERT, DAPK, RARβ, and CDH1) and their promoter methylation in HeLa (HPV18-positive), SiHa (HPV16-positive) and C33a (HPV-negative) cervical cancer cell lines. ALA increased DNA demethylase, HMTs, and HATs while decreasing global DNA methylation, DNMT, HDMs, and HDACs mRNA expression/activity in all cervical cancer cell lines. ALA downregulated hTERT oncogene while upregulating the mRNA expression of TSGs (Tumor Suppressor Genes) CDH1, RARβ, and DAPK in all the cell lines. ALA reduced methylation in the 5' CpG island of CDH1, RARβ, and DAPK1 promoters and reduced global DNA methylation in cervical cancer cell lines. These results suggest that ALA regulates the growth of cervical cancer cells by targeting epigenetic markers, shedding light on its potential therapeutic role in cervical cancer management.
Collapse
Affiliation(s)
- Amrita Ulhe
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
2
|
Vitek M, Zvonar Pobirk A, Roškar R, Matjaž MG. Exploiting the potential of in situ forming liquid crystals: development and in vitro performance of long-acting depots for peptide drug thymosin alpha 1 subcutaneous administration. Drug Deliv 2025; 32:2460708. [PMID: 40066714 PMCID: PMC11899226 DOI: 10.1080/10717544.2025.2460708] [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: 07/25/2024] [Revised: 11/06/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
The fast-growing filed of long-acting depots for subcutaneous (SC) administration holds significant potential to enhance patient adherence to treatment regimens, particularly in the context of chronic diseases. Among them, injectable in situ forming lyotropic liquid crystals (LCCs) consisting of hexagonal mesophases represent an attractive platform due to their remarkable highly ordered microstructure enabling the sustained drug release. These systems are especially relevant for peptide drugs, as their use is limited by their short plasma half-life and inherent poor stability. In this study, we thus aimed to exploit the potential of a liquid crystalline platform for the sustained release of peptide drug thymosin alpha 1 (Tα1), characterized by a short plasma half-life and with that associated twice-weekly SC administration regimen. We initially selected specified ingredients, with ethanol serving to reduce viscosity and stabilize the peptide drug Tα1, lecithin contributing to LCCs formation and stabilization, and glycerol monooleate or glycerol monolinoleate representing the hexagonal LCCs forming matrix material. The selected studied nonaqueous precursor formulations were characterized by suitable rheological properties for SC injection. A convenient and rapid in situ phase transition of precursor formulations to hexagonal LCCs, triggered by water absorption, was successfully accomplished in vitro. Notably, in situ formed LCCs demonstrated sustained release kinetics of the peptide drug Tα1 for up to 2 weeks of in vitro release testing, offering minimized dosing frequency and thus promoting patient adherence. In summary, the newly developed in situ forming liquid crystalline systems represent prospective injectable long-acting depots for SC administration of the peptide drug Tα1.
Collapse
Affiliation(s)
- Mercedes Vitek
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Alenka Zvonar Pobirk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Roškar
- Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Mirjam Gosenca Matjaž
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
3
|
Al-Beltagi M, Saeed NK, Bediwy AS, Alhawamdeh R, Elbeltagi R. Management of critical care emergencies in children with autism spectrum disorder. World J Crit Care Med 2025; 14:99975. [DOI: 10.5492/wjccm.v14.i2.99975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 12/13/2024] [Accepted: 12/30/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Managing critical care emergencies in children with autism spectrum disorder (ASD) presents unique challenges due to their distinct sensory sensitivities, communication difficulties, and behavioral issues. Effective strategies and protocols are essential for optimal care in these high-stress situations.
AIM To systematically evaluate and synthesize current evidence on best practices for managing critical care emergencies in children with ASD. The review focuses on key areas, including sensory-friendly environments, communication strategies, behavioral management, and the role of multidisciplinary approaches.
METHODS A comprehensive search was conducted across major medical databases, including PubMed, Embase, and Cochrane Library, for studies published between 2000 and 2023. Studies were selected based on their relevance to critical care management in children with ASD, encompassing randomized controlled trials, observational studies, qualitative research, and case studies. Data were extracted and analyzed to identify common themes, successful strategies, and areas for improvement.
RESULTS The review identified 50 studies that met the inclusion criteria. Findings highlighted the importance of creating sensory-friendly environments, utilizing effective communication strategies, and implementing individualized behavioral management plans. These findings, derived from a comprehensive review of current evidence, provide valuable insights into the best practices for managing critical care emergencies in children with ASD. Sensory modifications, such as reduced lighting and noise, visual aids, and augmentative and alternative communication tools, enhanced patient comfort and cooperation. The involvement of multidisciplinary teams was crucial in delivering holistic care. Case studies provided practical insights and underscored the need for continuous refinement of protocols.
CONCLUSION The review emphasizes the need for a tailored approach to managing critical care emergencies for children with ASD. Sensory-friendly adjustments, effective communication, and behavioral strategies supported by a multidisciplinary team are integral to improving outcomes. Despite progress, ongoing refinement of care practices and protocols is necessary. This ongoing process addresses remaining challenges and engages healthcare professionals in continuous improvement of care for children with ASD in critical settings.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Manama, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 26671, Manama, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Manama, Bahrain
| | - Rawan Alhawamdeh
- Department of Pediatrics Research and Development, Sensoryme Dwc-llc, Dubai 712495, Dubai, United Arab Emirates
- Department of Pediatrics Research and Development, Genomics Sensory Play and Creativity Center, Manama 22673, Manama, Bahrain
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland-Bahrain, Busiateen 15503, Muharraq, Bahrain
| |
Collapse
|
4
|
Wei Y, Zheng F, Guo L, Chen W, Wang H, Bao L, Wu Z, Li Y, Alateng W, Su J, Kong M, Ke S. Natural pyrrolo[1,2-a]quinazolinone derivatives: Design, synthesis, characterization, and bio-evaluation as novel antiviral agents. Eur J Med Chem 2025; 288:117383. [PMID: 39978108 DOI: 10.1016/j.ejmech.2025.117383] [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: 12/17/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
As viral infectious diseases increasingly threaten global health, antiviral drug research has become a focus in the medicinal chemistry. Enterovirus has always been an important virus causing infections disease with a high incidence in summer and autumn, such as Enterovirus 71 (EV71) and Coxsackievirus B3 (CVB3). Currently, no specific antiviral drugs are available for EV71 and CVB3. So, we designed and synthesized a novel series of quinazolinone derivatives based on the natural pyrrolo[1,2-a]quinazolinone scaffold, which were fully characterized and identified as potential anti-enterovirus agents. Among them, compound B9 exhibited potent anti-CVB3 activity with an EC50 value of 17.4 ± 3.62 μM, and compound B5 exhibited potent anti-EV71 activity with an EC50 value of 14.8 ± 2.18 μM as confirmed by determining the cytopathic effects, progeny virus titers, viral nucleic acid and protein levels. The potential antiviral mechanisms of compound B5 were also explored. The compound B5 exhibited a powerful therapeutic effect primarily by blocking the post-attachment stage of viral infection. Further experiments demonstrated that compound B5 didn't inhibit the activities of the EV71 2Apro and 3Dpol. Modelling of the molecular binding of the 3Cpro-compound complex revealed that the compound B5 could insert into the substrate-binding pocket of EV71 3Cpro, blocking substrate recognition and possibly inhibiting EV71 3Cpro activity. These researches may provide evidence for the development of these novel pyrrolo[1,2-a]quinazolinone derivatives derived from natural products as potential antiviral agents.
Collapse
Affiliation(s)
- Yanhong Wei
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Fuqiang Zheng
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China; Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Lirong Guo
- Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; College of Chemistry, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenxuan Chen
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Hexiang Wang
- Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; College of Chemistry, Huazhong Agricultural University, Wuhan, 430070, China
| | - Longzhu Bao
- Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Zhaoyuan Wu
- Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Ying Li
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - W Alateng
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Jianglong Su
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Mengmeng Kong
- Sino-German Biomedical Center, School of Life and Health Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China
| | - Shaoyong Ke
- Key Lab of Microbial Pesticides (Ministry of Agriculture and Rural Affairs), National Biopesticide Engineering Research Centre, Hubei Biopesticide Engineering Research Centre, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China.
| |
Collapse
|
5
|
Baradaran B, Hazrati A, Kazemi-Sefat NA, Soleimanjahi H, Soudi S. Umbilical cord-derived mesenchymal stem cell condition medium effect on rotavirus-infected Caco-2 cells survival and inflammatory responses. Tissue Cell 2025; 93:102699. [PMID: 39818065 DOI: 10.1016/j.tice.2024.102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
Rotavirus is the most important cause of severe gastroenteritis in infants and children worldwide. This virus causes an increase in inflammatory responses by increasing cellular oxidative stress and the expression and activity of the transcription factor NF-κB and COX-2. As a result of NF-κB activation, the expression of inflammatory cytokines also increases. So, there is a need to control pathogenic inflammatory responses mediated by rotavirus. Mesenchymal stem cells (MSCs) have confirmed immunomodulatory characteristics. The present study aims to investigate the effects of MSCs conditioned media (MSCs-CM) in reducing the inflammatory response of Caco-2 cells when exposed to rotavirus. 72 h After rotavirus-infected Caco-2 cell of treatment with MSCs-CM, virus replication (CCID50), secretion of IL-6, and IL-8 (ELISA), COX-2 and NF-κB genes expression (q-PCR), apoptosis (Annexin V-PI), and nitric oxide (NO) level (Gries's reagent) are investigated. Based on the results, virus replication was reduced by Log1 in the CM-treated groups. Also, treating Caco-2 cells with MSCs-CM led to decreased IL-6 and NO and increased IL-8 production. Evaluation of apoptosis in MSCs-CM-treated rotavirus-exposed Caco-2 cells showed a significant reduction in their apoptosis. Also, the expression of COX-2 is increased significantly. However, the expression of NF-κB decreased significantly after treatment with MSCs-CM. The results show that inflammatory responses, oxidative stress, and apoptosis in rotavirus-infected cells have decreased after treatment with MSC-CM.
Collapse
Affiliation(s)
- Behnoosh Baradaran
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
6
|
Lu W, Yang X, Wang B. Carbon monoxide potentiates the effect of corticosteroids in suppressing inflammatory responses in cell culture. Bioorg Med Chem 2025; 120:118092. [PMID: 39904198 DOI: 10.1016/j.bmc.2025.118092] [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: 11/02/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Inflammation is a pathology implicated in a wide range of human diseases. Recent years have seen tremendous progress in developing new types of anti-inflammatory agents for the treatment of inflammation of various origins. However, each has its own strengths and weaknesses. The very fact that there needs to have multiple types of anti-inflammatory agents underlines the complexity of inflammatory diseases and conditions, their molecular origins, and their treatment. Such complexity dictates the need to search for new approaches with improved potency and efficacy as well as reduced side effects. For these reasons, we are interested in exploring the possibility of generating synergy between carbon monoxide (CO), an endogenously produced cytoprotective agent, and known anti-inflammatory agents. Herein, we report the potentiating actions of CO on the anti-inflammatory effects of cortisone and dexamethasone as demonstrated in their ability to suppress the expression of TNF-α and IL-6 induced by either LPS or the S protein of SARS-CoV-2. Such effects are reflected in the substantially increased potency as well efficacy, when the efficacy of the corticosteroid alone does not allow for complete suppression of the expression of these cytokines. Further, increased attenuation of p65 phosphorylation is at least part of the molecular mechanism for the observed potentiating effects. We hope our work will stimulate a high level of activity along the same direction, leading to anti-inflammatory strategies with improved potency and efficacy and reduced side effects.
Collapse
Affiliation(s)
- Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
7
|
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
|
8
|
Nagoba BS, Dhotre SV, Gavkare AM, Mumbre SS, Dhotre PS. Convergence of COVID-19 and recurrent stroke: In-hospital mortality risks explored. World J Virol 2025; 14:99904. [DOI: 10.5501/wjv.v14.i1.99904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
This editorial comments on the article by Desai et al, which investigates the impact of coronavirus disease 2019 (COVID-19) on in-hospital mortality among patients with recurrent stroke using data from the 2020 National Inpatient Sample. The findings reveal significantly higher mortality rates in COVID-19-positive patients compared to non-COVID-19 patients, particularly among middle-aged individuals, males, and ethnic minorities. This editorial explores the underlying mechanisms contributing to these outcomes and discusses the clinical implications for targeted management strategies in high-risk groups. The results emphasize the need for comprehensive approaches to mitigate the heightened risks faced by recurrent stroke patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Basavraj S Nagoba
- Department of Microbiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Shree V Dhotre
- Department of Microbiology, Ashwini Rural Medical College, Solapur 413001, India
| | - Ajay M Gavkare
- Department of Physiology, Maharashtra Institute of Medical Sciences and Research (Medical College), Latur 413531, India
| | - Sachin S Mumbre
- Department of Community Medicine, Ashwini Rural Medical College, Solapur 413001, India
| | - Pradnya S Dhotre
- Department of Biochemistry, Ashwini Rural Medical College, Solapur 413001, India
| |
Collapse
|
9
|
Shamim L, Musharaf I, Nashwan AJ. Dexamethasone in coronavirus disease 2019 care: Dosage and utilization insights. World J Virol 2025; 14:98765. [DOI: 10.5501/wjv.v14.i1.98765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2. It was declared a global pandemic on March 11, 2020, by the World Health Organization. An excessive inflammatory response is a severe respiratory manifestation of COVID-19, which becomes predominant in later stages. Due to its immunosuppressive and anti-inflammatory properties, dexamethasone is the first systemic glucocorticoid to treat severe COVID-19 patients. This editorial reviews the efficacy and safety of high-dose vs low-dose dexamethasone in patients with COVID-19. Findings indicate that using low-dose dexamethasone is beneficial and emphasize the need for additional research on the use of high-dose dexamethasone. While the study provides a robust evidence base, it is limited by the lack of long-term data, focus on specific outcomes and heterogeneity of the included studies. Future research should focus on the long-term effects of dexamethasone and its impact across varying disease severities and patient populations to refine treatment strategies and improve patient care.
Collapse
Affiliation(s)
- Laiba Shamim
- Department of Clinical Medicine, Jinnah Sindh Medical University, Karachi 75510, Sindh, Pakistan
| | - Imshaal Musharaf
- Department of Medicine, Jinnah Sindh Medical University, Karachi 75510, Sindh, Pakistan
| | - Abdulqadir J Nashwan
- Department of Nursing and Midwifery Research, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
10
|
Varama A. Revisiting dexamethasone dosage in COVID-19 management. World J Virol 2025; 14:98359. [DOI: 10.5501/wjv.v14.i1.98359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has necessitated rapid advancements in therapeutic strategies, with dexamethasone emerging as a key treatment for severe cases. This editorial discusses the systematic review conducted by Sethi et al, published in the World Journal of Virology. The review critically examines the efficacy and safety of varying dosages of dexamethasone in severe COVID-19 patients, providing a comprehensive meta-analysis that underscores the current clinical recommendations favoring a low-dose regimen. Despite these findings, the review highlights the potential benefits of tailored dosages for specific patient subgroups, suggesting a need for personalized treatment approaches. This editorial expands on the implications of these findings, advocating for the integration of evolving clinical data into treatment protocols and calling for further research into patient-specific responses to therapy. It emphasizes the importance of adaptability and precision in pandemic response, urging the medical community to consider both the robustness of existing evidence and the potential for innovative approaches to enhance patient outcomes in the face of global health challenges.
Collapse
Affiliation(s)
- Abhishet Varama
- Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Krung Thep Maha Nakhon, Thailand
| |
Collapse
|
11
|
Sarker MS. Rhabdomyolysis-related acute kidney injury in COVID-19: A critical concern. World J Virol 2025; 14:100160. [DOI: 10.5501/wjv.v14.i1.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/24/2024] [Accepted: 10/15/2024] [Indexed: 11/28/2024] Open
Abstract
Rhabdomyolysis is a severe condition characterized by the breakdown of muscle tissue leading to the release of intracellular components into the bloodstream. This condition, when associated with acute kidney injury (AKI), can result in significant morbidity and mortality, particularly in the context of coronavirus disease 2019 (COVID-19). This editorial discusses a retrospective study on patients with COVID-19 who developed rhabdomyolysis-related AKI. The study highlights that patients with rhabdomyolysis exhibited higher inflammatory markers, such as C-reactive protein, ferritin, and procalcitonin, and experienced worse clinical outcomes compared to those with other causes of AKI. The findings underscore the importance of early recognition and management of rhabdomyolysis in COVID-19 patients to improve prognosis and reduce mortality rates.
Collapse
|
12
|
Saeed NK, Almusawi SK, Albalooshi NA, Al-Beltagi M. Unveiling the impact: COVID-19's influence on bacterial resistance in the Kingdom of Bahrain. World J Virol 2025; 14:100501. [DOI: 10.5501/wjv.v14.i1.100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/22/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Antibiotic resistance is a growing global health threat, and understanding local trends in bacterial isolates and their susceptibility patterns is crucial for effective infection control and antimicrobial stewardship. The coronavirus disease 2019 (COVID-19) pandemic has introduced additional complexities, potentially influencing these patterns.
AIM To analyze trends in bacterial isolates and their antibiotic susceptibility patterns at Salmaniya Medical Complex from 2018 to 2023, with a specific focus on the impact of the COVID-19 pandemic on these trends.
METHODS A retrospective analysis of microbiological data was conducted, covering the period from 2018 to 2023. The study included key bacterial pathogens such as Escherichia coli (E. coli), Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Staphylococcus aureus, among others. The antibiotic susceptibility profiles of these isolates were assessed using standard laboratory methods. To contextualize the findings, the findings were compared with similar studies from other regions, including China, India, Romania, Saudi Arabia, the United Arab Emirates, Malaysia, and United States.
RESULTS The study revealed fluctuating trends in the prevalence of bacterial isolates, with notable changes during the COVID-19 pandemic. For example, a significant increase in the prevalence of Staphylococcus aureus was observed during the pandemic years, while the prevalence of E. coli showed a more variable pattern. Antibiotic resistance rates varied among the different pathogens, with a concerning rise in resistance to commonly used antibiotics, particularly among Klebsiella pneumoniae and E. coli. Additionally, the study identified an alarming increase in the prevalence of multidrug-resistant (MDR) strains, especially within Klebsiella pneumoniae and E. coli isolates. The impact of the COVID-19 pandemic on these trends was evident, with shifts in the frequency, resistance patterns, and the emergence of MDR bacteria among several key pathogens.
CONCLUSION This study highlights the dynamic nature of bacterial isolates and their antibiotic susceptibility patterns at Salmaniya Medical Complex, particularly in the context of the COVID-19 pandemic. The findings underscore the need for continuous monitoring and effective anti-microbial stewardship programs to combat the evolving threat of antibiotic resistance. Further research and policy initiatives are required to address the identified challenges and improve patient outcomes in the face of these ongoing challenges.
Collapse
Affiliation(s)
- Nermin K Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Governmental Hospitals, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Royal College of Surgeons in Ireland–Medical University of Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Safiya K Almusawi
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Governmental Hospitals, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Royal College of Surgeons in Ireland–Medical University of Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Noor A Albalooshi
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Governmental Hospitals, Manama 12, Bahrain
| | - Mohammed Al-Beltagi
- Department of Paediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Algharbia, Bahrain
| |
Collapse
|
13
|
Kalamvoki M. HSV-1 virions and related particles: biogenesis and implications in the infection. J Virol 2025; 99:e0107624. [PMID: 39898651 DOI: 10.1128/jvi.01076-24] [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] [Indexed: 02/04/2025] Open
Abstract
Virion formation and egress are sophisticated processes that rely on the spatial and temporal organization of host cell membranes and the manipulation of host machineries involved in protein sorting, membrane bending, fusion, and fission. These processes result in the formation of infectious virions, defective particles, and various vesicle-like structures. In herpes simplex virus 1 (HSV-1) infections, virions and capsid-less particles, known as light (L)-particles, are formed. HSV-1 infection also stimulates the release of particles that resemble extracellular vesicles (EVs). In productively infected cells, most EVs are generated through the CD63 tetraspanin biogenesis pathway and lack viral components. A smaller subset of EVs, generated through the endosomal sorting complexes required for transport (ESCRT) pathway, contains both viral and host factors. Viral mechanisms tightly regulate EV biogenesis, including the inhibition of autophagy-a process critical for increased production of CD63+ EVs during HSV-1 infection. Mutant viruses that fail to suppress autophagy instead promote microvesicle production from the plasma membrane. Additionally, the viral protein ICP0 (Infected Cell Protein 0) enhances EV biogenesis during HSV-1 infection. The different types of particles can be separated by density gradients due to their distinct biophysical properties. L-particles and ESCRT+ EVs display a pro-viral role, supporting viral replication, whereas CD63+ EVs exhibit antiviral effects. Overall, these studies highlight that HSV-1 infection yields numerous and diverse particles, with their type and composition shaped by the ability of the virus to evade host responses. These particles likely shape the infectious microenvironment and determine disease outcomes.
Collapse
Affiliation(s)
- Maria Kalamvoki
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
14
|
Eke AC, Brummel SS, Aliyu MH, Stranix-Chibanda L, Eleje GU, Ezebialu IU, Korutaro V, Wabwire D, Matubu A, Mbengeranwa T, Chakhtoura N, Chinula L, McCarthy K, Knowles K, Krotje C, Linton MF, Dooley KE, Sax PE, Brown T, Lockman S. Lipid and Glucose Profiles in Pregnant Women With HIV on Tenofovir-based Antiretroviral Therapy. Clin Infect Dis 2025; 80:594-601. [PMID: 39219495 PMCID: PMC11912963 DOI: 10.1093/cid/ciae441] [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: 06/03/2024] [Revised: 07/23/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Tenofovir alafenamide (TAF)-based antiretroviral therapy (ART) regimens have been associated with adverse changes in lipid and glucose profiles compared with tenofovir disoproxil fumarate (TDF)-based ART, but data in pregnancy are limited. We evaluated metabolic markers in pregnant women with human immunodeficiency virus (HIV) after starting TAF- versus TDF-based ART. METHODS We analyzed data within the IMPAACT 2010/VESTED trial, which demonstrated better pregnancy outcomes in pregnant women randomized to initiate TAF/Emtricitabine/Dolutegravir (TAF/FTC + DTG; n = 217) or TDF/FTC + DTG (n = 215). We measured non-fasting plasma concentrations of glucose, total-cholesterol, low-density lipoprotein-cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), lipoprotein (a), and triglycerides from samples collected 8 weeks after enrollment. We employed linear regression models to estimate by-arm mean differences. RESULTS In total, 219 participants enrolled in the DTG arms in Zimbabwe and Uganda: 109 in the TAF/FTC + DTG and 110 in the TDF/FTC + DTG arms. At study entry, mean gestational age was 22.6 weeks, median HIV-1 RNA was 711 copies/mL, and mean age was 25.8 years. By 8 weeks, mean total cholesterol was 12 mg/dL higher in women randomized to TAF/ FTC + DTG versus TDF/FTC + DTG (95% confidence interval [CI]: 3.8, 21.1). Pregnant women in the TAF/FTC + DTG arm had higher mean LDL-C (7.1 mg/dL, 95% CI: .2, 14.0), triglycerides (12.3 mg/dL, 95% CI: 1.8, 22.7), lipoprotein (a) (7.3 mg/dL, 95% CI: 1.1, 13.6), and lower mean HDL-C (2.8 mg/dL, 95% CI: .1, 5.6) compared to the TDF/FTC + DTG arm. CONCLUSIONS Pregnant women randomized to start TAF/FTC + DTG had higher lipids than those randomized to TDF/FTC + DTG within 8 weeks of ART initiation. However, lipid levels were within normal reference ranges.
Collapse
Affiliation(s)
- Ahizechukwu C Eke
- Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sean S Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Muktar H Aliyu
- Department of Health Policy and Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lynda Stranix-Chibanda
- Child and Adolescent Health Unit, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
- Faculty of Medicine and Health Sciences, University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - George U Eleje
- Department of Obstetrics and Gynecology, Nnamdi Azikiwe University Teaching Hospital, Nnewi, Anambra State, Nigeria
| | - Ifeanyichukwu U Ezebialu
- Department of Obstetrics and Gynecology, Chukwuemeka Odumegwu Ojukwu University Teaching Hospital, Amaku, Anambra State, Nigeria
| | - Violet Korutaro
- Baylor College of Medicine Children's Foundation Uganda, Kampala, Uganda
| | - Deo Wabwire
- Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Allen Matubu
- Faculty of Medicine and Health Sciences, University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Tapiwa Mbengeranwa
- Faculty of Medicine and Health Sciences, University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Nahida Chakhtoura
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Lameck Chinula
- Division of Global Women's Health, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Obstetrics and Gynecology, UNC Project Malawi, Lilongwe, Malawi
| | | | | | | | - Macrae F Linton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelly E Dooley
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Paul E Sax
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Todd Brown
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shahin Lockman
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Botswana Harvard Health Partnership, Gaborone, Botswana
| |
Collapse
|
15
|
Moslehi S, Narimani S, Soleimanpour S. Challenges and Adaptive Measures for the Potential Next Pandemic Caused by Climate Change. Disaster Med Public Health Prep 2025; 19:e59. [PMID: 40091659 DOI: 10.1017/dmp.2025.56] [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: 03/19/2025]
Abstract
Climate change is increasingly recognized as a significant driver of emerging infectious diseases, with the potential to catalyze the next global pandemic. This paper explores the complex interplay between climate change and the emergence of novel pathogens, emphasizing the environmental, ecological, and socio-economic factors that contribute to disease transmission. Rising temperatures, altered precipitation patterns, and habitat destruction are reshaping ecosystems, bringing humans into closer contact with zoonotic reservoirs and vectors. These changes amplify the risk of spillover events, as seen in recent outbreaks. The study identifies key challenges, including inadequate surveillance systems, limited global cooperation, and the disproportionate impact on vulnerable populations. Furthermore, it proposes adaptive measures such as enhanced early warning systems, integrated One Health approaches, and climate-resilient public health infrastructure. By addressing these challenges and implementing proactive strategies, the global community can mitigate the risk of a climate-driven pandemic and strengthen preparedness for future health crises. This paper underscores the urgent need for interdisciplinary collaboration and policy innovation to safeguard global health in the face of a changing climate.
Collapse
Affiliation(s)
- Shandiz Moslehi
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
- Department of Health in Disasters and Emergencies, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Narimani
- Department of Health in Disasters and Emergencies, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
- Department of Nursing and midwifery, School of Nursing, Social Determinant of Health Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Samira Soleimanpour
- Assistant professor of Medical Librarianship and information sciences, Educational Development Center (EDC), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Demirkaya Miloglu F, Bayrak B, Yuksel B, Demir SN, Gundogdu G, Kadioglu Y, Abd El-Aty AM. Plasma metabolomic signatures after oral administration of ritonavir in COVID-19 treatment via chemometrics-assisted UPLC/Q-TOF/MS/MS. J Pharm Biomed Anal 2025; 255:116638. [PMID: 39700866 DOI: 10.1016/j.jpba.2024.116638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/02/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Understanding the pharmacodynamics of ritonavir through metabolomics offers insights into its side effects and helps in the development of safer therapies. This study aimed to investigate the effects of ritonavir treatment on the metabolic profiles of rabbits via a metabolomics approach, with the objective of elucidating its impact on various biochemical pathways and identifying relevant biomarkers. The rabbits were divided into control and ritonavir-treated groups, and their plasma samples were analyzed via ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF/MS/MS). Metabolites were identified on the basis of the masscharge ratio (m/z) and validated via XCMS software. Metabolites with a fold change ≥ 1.5 and P ≤ 0.01 were analyzed via principal component analysis (PCA) and orthogonal partial least squares discrimination analysis (OPLS-DA) to distinguish between the groups. MetaboAnalyst 6.0 was used for pathway analysis to identify metabolic pathways affected by ritonavir. The PCA and OPLS-DA models revealed clear separation between the control and ritonavir-treated groups, with high R² and Q² values indicating robust model performance. Pathway analysis revealed that ritonavir treatment significantly affected several metabolic pathways, including those related to ether lipid, phenylalanine, sphingolipid, and glycerophospholipid metabolism. Particularly significant changes were observed in metabolites related to lipid metabolism, oxidative stress responses and cellular signaling. Ritonavir significantly impacts metabolic pathways, particularly those involved in lipid metabolism, and oxidative stress responses, which may influence immune responses and drug interactions. This study also highlights the potential of integrating metabolomics with personalized medicine approaches to optimize ritonavir treatment strategies and reduce adverse effects. These findings indicate that ritonavir significantly influences cellular homeostasis and metabolic processes in addition to its antiviral properties. This highlights the necessity of comprehending the metabolic effects of ritonavir to enhance its clinical application, especially in the management of COVID-19. Further research is warranted to explore these alterations and their implications for therapeutic strategies.
Collapse
Affiliation(s)
- Fatma Demirkaya Miloglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey.
| | - Burak Bayrak
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Busra Yuksel
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Sema Nur Demir
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Gulsah Gundogdu
- Department of Physiology, Faculty of Medicine, Pamukkale University, Erzurum 20070, Turkey
| | - Yucel Kadioglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey.
| |
Collapse
|
17
|
Sanchez BC, Ortiz RM, Grasis JA. Human adenovirus serotype 5 infection dysregulates cysteine, purine, and unsaturated fatty acid metabolism in fibroblasts. FASEB J 2025; 39:e70411. [PMID: 40052831 PMCID: PMC11887610 DOI: 10.1096/fj.202402726r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/30/2025] [Accepted: 02/12/2025] [Indexed: 03/09/2025]
Abstract
Viral infections can cause cellular dysregulation of metabolic reactions. Viruses alter host metabolism to meet their replication needs. The impact of viruses on specific metabolic pathways is not well understood, even in well-studied viruses, such as human adenovirus. Adenoviral infection is known to influence cellular glycolysis and respiration; however, global effects on overall cellular metabolism in response to infection are unclear. Furthermore, few studies have employed an untargeted approach, combining emphasis on viral dosage and infection. To address this, we employed untargeted metabolomics to quantify the dynamic metabolic shifts in fibroblasts infected with human adenovirus serotype 5 (HAdV-5) at three dosages (0.5, 1.0, and 2.0 multiplicity of infection [MOI]) and across 4 time points (6-, 12-, 24-, and 36-h post-infection [HPI]). The greatest differences in individual metabolites were observed at 6- and 12-h post-infection, correlating with the early phase of the HAdV-5 infection cycle. In addition to its effects on glycolysis and respiration, adenoviral infection downregulates cysteine and unsaturated fatty acid metabolism while upregulating aspects of purine metabolism. These results reveal specific metabolic pathways dysregulated by adenoviral infection and the associated dynamic shifts in metabolism, suggesting that viral infections alter energetics via profound changes in lipid, nucleic acid, and protein metabolism. The results revealed previously unconsidered metabolic pathways disrupted by HAdV-5 that can alter cellular metabolism, thereby prompting further investigation into HAdV mechanisms and antiviral targeting.
Collapse
Affiliation(s)
- Bailey‐J C. Sanchez
- Quantitative and Systems Biology, School of Natural SciencesUniversity of CaliforniaMercedCaliforniaUSA
| | - Rudy M. Ortiz
- Quantitative and Systems Biology, School of Natural SciencesUniversity of CaliforniaMercedCaliforniaUSA
| | - Juris A. Grasis
- Quantitative and Systems Biology, School of Natural SciencesUniversity of CaliforniaMercedCaliforniaUSA
| |
Collapse
|
18
|
Scalzo PL, Marshall AG, Soriano S, Curry K, Dulay M, Hodics T, Quigley EMM, Treangen TJ, Piskorz MM, Villapol S. Gut Microbiome dysbiosis and immune activation correlate with somatic and neuropsychiatric symptoms in COVID-19 patients. J Transl Med 2025; 23:327. [PMID: 40087795 PMCID: PMC11907868 DOI: 10.1186/s12967-025-06348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Infection with SARS-CoV-2, the virus responsible for COVID-19, can lead to a range of physical symptoms and mental health challenges, including stress, anxiety, and depression. These effects are particularly pronounced in hospitalized patients, likely due to the virus's direct and indirect impact on the nervous system. Gut dysbiosis, an imbalance in the gut microbiome, has been implicated in immune dysfunction and chronic inflammation in COVID-19 patients. However, the interactions between gut microbiome composition and the physical and mental symptoms of COVID-19 remain incompletely understood. METHODS We investigated the association between physical and mental symptoms, cytokine profiles, and gut microbiota composition in 124 hospitalized COVID-19 patients. We collected data on demographics, COVID-19 severity, and mental health indicators (stress, anxiety, and depression). Gut microbiome profiling was performed using full-length 16 S rRNA gene sequencing to evaluate microbial diversity and composition. RESULTS COVID-19 severity was categorized as low (27.4%), moderate (29.8%), or critical (42.8%). Common symptoms included fever (66.1%) and cough (55.6%), while somatic symptoms (27.3%), anxiety (27.3%), depressive symptoms (39%), and stress (80.5%) were frequently self-reported. Elevated interleukin-6 levels in severe cases highlighted systemic inflammation, reduced gut bacterial diversity, particularly among women and obese patients, correlated with higher disease severity. Notably, the genus Mitsuokella was associated with increased physical symptoms and mental distress, while Granulicatella was linked to critical illness. CONCLUSIONS Our findings reveal significant associations between mental health status, systemic inflammation, and gut dysbiosis in hospitalized COVID-19 patients. These results indicate the potential for microbiome-targeted therapies to mitigate psychological and physical complications and improve recovery outcomes in this population.
Collapse
Affiliation(s)
- Paula L Scalzo
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Austin G Marshall
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Kristen Curry
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mario Dulay
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Timea Hodics
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Eamonn M M Quigley
- Lynda K. and David M. Underwood Center for Digestive Health, Houston Methodist Hospital, Houston, TX, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - María M Piskorz
- Department of Neurogastroenterology, Hospital de Clinicas José de San Martin, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sonia Villapol
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
19
|
Feng C, Shi J, Chen Y, Chen S, Cui J, Zhang J, Zheng X, Wang Y, Li F. A hepatitis B virus-free cccDNA-producing stable cell for antiviral screening. Antiviral Res 2025:106143. [PMID: 40090467 DOI: 10.1016/j.antiviral.2025.106143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
The covalently closed circular DNA (cccDNA) of the Hepatitis B virus (HBV) serves as a template for producing progeny viruses in virally infected hepatocytes. Promising cccDNA-targeting antiviral agents remain unavailable and unpredictable in the research and development pipelines, making sterile HBV elimination challenging at the current stage. The major challenge of discriminating trace amounts of cccDNA from the abundant HBV relaxed circular DNA (rcDNA), which is nearly identical to cccDNA in sequence, substantially discourages efforts to discover and directly screen cccDNA-targeting drugs. Therefore, an easy cccDNA cell culture system is required for high-throughput drug screening. In this study, we designed an HBV cccDNA self-generating stable cell culture system using a functional complementary concept and successfully generated an HBV cccDNA Gaussia luciferase reporter cell line in HepG2 and Huh7 cells. This design ensures that the Gluc signal is exclusively expressed upon cccDNA formation, allowing for the accurate and easy measurement of cccDNA levels via luminescent signals. Using this system, in conjunction with a firefly luciferase reporter to monitor cell activity, we screened 2,074 drugs in the HepG2-HBV-cccDNA/Firefly cell line. Four compounds were selected for further experimentation and their anti-HBV effects were confirmed. Thus, this virus-free hepatitis B cccDNA cell culture system provides a valuable and convenient platform for the high-throughput screening of anti-HBV drugs.
Collapse
Affiliation(s)
- Chengqian Feng
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Jingrong Shi
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Yunfu Chen
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co., Ltd, Dongguan 523871, China
| | - Sisi Chen
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Jianping Cui
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Jun Zhang
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Xiaowen Zheng
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Yaping Wang
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China
| | - Feng Li
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China, 8 Huaying Road, Baiyun District, Guangzhou, Guangdong Province, 510663, China; Scientific Research Center, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| |
Collapse
|
20
|
Vetsika EK, Katsianou MA, Sarantis P, Palamaris K, Papavassiliou AG, Piperi C. Pediatric gliomas immunity challenges and immunotherapy advances. Cancer Lett 2025:217640. [PMID: 40090572 DOI: 10.1016/j.canlet.2025.217640] [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: 02/11/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Pediatric gliomas, the most frequent brain tumors in children, are characterized by heterogeneity and a unique tumor immune microenvironment. They are categorized into different subtypes, including low-grade gliomas like pilocytic astrocytomas and high-grade gliomas such as diffuse midline gliomas and diffuse intrinsic pontine gliomas, each exhibiting distinct immunological profiles. The tumor immune microenvironment in pediatric gliomas is shaped by cellular and non-cellular components, including immune cells, cytokines, and the extracellular matrix, involved in tumor progression, immune evasion, and response to therapy. While pediatric low-grade gliomas often display an immunosuppressed microenvironment, high-grade gliomas are characterized by complex immune infiltrates and intricate immunosuppressive mechanisms. The blood-brain barrier further obscures immune cell recruitment and therapeutic delivery. Despite advances in understanding adult gliomas, the immunobiology of pediatric tumors is poorly investigated, with limited data on the interactions between glioma cells and immune populations such as T and natural killer cells, as well as tumor-associated macrophages. Herein, we provide an update of the current knowledge on tumor immune microenvironment interactions in pediatric gliomas, highlighting the immunosuppressive mechanisms and emerging immunotherapeutic strategies aiming at overcoming these barriers to improve clinical outcomes for affected children.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria A Katsianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Palamaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
21
|
He P, He C, Wu F, Ou Y, Luo S, Zhang Y, Chang Y, Guo Z, Tang X, Zhao Y, Xu Y, Wang H, Bai S, Du G, Sun X. Microneedle-delivered adeno-associated virus vaccine amplified anti-viral immunity by improving antigen-presenting cells infection. J Control Release 2025; 379:1045-1057. [PMID: 39875077 DOI: 10.1016/j.jconrel.2025.01.069] [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: 11/20/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Adeno-associated viruses (AAV) have significant potential as vaccine carriers due to their excellent biosafety and efficient antigen gene delivery. However, most AAV vaccines show limited capacity to transduce antigen-presenting cells (APCs) following intramuscular injection which may cause inadequate cellular immune responses and undesired side effects due to transducing other tissues or cells. Herein, we developed a soluble microneedle patch for targeting the AAV vaccines to the epidermal and dermal APCs. To preserve the biological activity of the AAV vaccine, the microneedles were fabricated via an optimized two-step low-temperature strategy and using 20 % trehalose as a protective agent. AAV serotype 8, which expresses the trimeric receptor-binding domain (RBD) of the SARS-CoV-2 spike protein (AAV8-RBD), remained 100 % biological activity after being loaded into the microneedles (MN-A8R). Upon a single-dose vaccination on the dorsal skin of mice, MN-A8R efficiently recruited APCs to the vaccination site and improved AAV8-RBD infection in APCs. Furthermore, MN-A8R prompted an increased formation of germinal centers in the draining lymph nodes. Compared to hypodermic needle-mediated intradermal injection, MN-A8R induced significantly stronger cellular immune responses and long-lasting, high-quality neutralizing antibodies. Importantly, MN-A8R demonstrated more comprehensive and robust cross-protection against three common SARS-CoV-2 pseudoviruses for at least six months. Our findings highlight the use of optimized polymeric microneedles for preserving AAV vaccine biological activity and enhancing the AAV vaccine efficacy by up-regulating APC infection.
Collapse
Affiliation(s)
- Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fuhua Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuang Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yongshun Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yu Chang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhaofei Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuanhao Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuting Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
22
|
Kordi R, Chang AJ, Hicar MD. Serology supportive of recent coxsackievirus B infection is correlated with multisystem inflammatory syndrome in children (MIS-C). Microbiol Spectr 2025; 13:e0174124. [PMID: 39907434 DOI: 10.1128/spectrum.01741-24] [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: 07/15/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Rarely, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will lead to myocarditis associated with multisystem inflammatory syndrome in children (MIS-C). It remains unclear why MIS-C only targets specific children. To explore an association between coxsackievirus infections with MIS-C, we investigated the sero-epidemiology of CV in admitted pediatric patients in relation to the pandemic. This retrospective case-control study was performed by chart review of children (age ≤21 years) admitted to a tertiary care hospital with CV serological testing from January 2017 to August 2023. Clinical, laboratory, and imaging findings were used to classify patients as MIS-C and CV-unlikely or CV-possible for non-MIS-C patients. Out of 182 admissions (179 patients, median age, 6), CVB complement fixation (CF) assay on serotypes B1-B6 and CVA immunofluorescence assay IgG on serotypes A7, A9, A16, and A24 were positive in at least one serotype in 59.2% and 80.7% of cases, respectively. We observed a significant drop in CVB CF seropositivity during the peak of social distancing in 2020. The likelihood of elevated CVB CF titers was significantly higher in MIS-C than the CV-unlikely group (OR: 1.92, 95% CI: 1.02-3.63, P: 0.04) and showed a trend toward higher values in African Americans than Whites (OR: 1.57, 95% CI: 0.98-2.50, P: 0.057). The frequency of MIS-C was considerably higher in African Americans than Whites (18.1% versus 9%, P: 0.1). A higher likelihood of elevated CVB CF titers in patients with MIS-C compared with those unlikely to have acute CV infection along with a relatively higher frequency of MIS-C in African Americans warrants further investigation into the role of CVB infection in MIS-C development.IMPORTANCEThe emergence of multisystem inflammatory syndrome in children (MIS-C) during the SARS-CoV-2 pandemic raised major concerns in providers caring for children. This condition presents a hyper-inflammation state that can lead to severe complications, including myocarditis and cardiogenic shock. The pathogenesis of MIS-C has not been fully understood. Understanding the pathogenesis of this condition is not only important for developing effective treatments but also for applying preventive strategies. A two-hit hypothesis leading to MIS-C has been proposed. Coxsackievirus infections are prevalent during childhood and can also cause myocarditis, and coxsackievirus B specifically has been shown to cause persistent RNA presence in host cells, leading to continued inflammation. Herein, we show that elevated coxsackievirus B titers are associated with MIS-C cases, implying a role of successive infections with these viruses contributing to such a hyperinflammatory state. This study supports the need for larger investigations into this association.
Collapse
Affiliation(s)
- Ramesh Kordi
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, New York, USA
| | - Arthur J Chang
- Division of Pediatric Infectious Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mark D Hicar
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
23
|
Rayle MR, Brunner JL, Dahrouge NC, Keller EL, Rittenhouse TAG. Infection intensity and severity of Ranavirus transmission in juvenile wood frogs. Oecologia 2025; 207:46. [PMID: 40035880 PMCID: PMC11880146 DOI: 10.1007/s00442-025-05682-8] [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: 03/23/2024] [Accepted: 02/15/2025] [Indexed: 03/06/2025]
Abstract
Ranaviruses are responsible for mass die offs of wood frog (Lithobates sylvaticus) tadpoles. What happens in between epidemics is less clear, but juvenile (metamorphosed) stages are hypothesized to move Ranaviruses among wetlands and introduce or reintroduce these viruses into wetlands, initiating new outbreaks. A key question is under what circumstances can juvenile L. sylvaticus infect susceptible conspecifics. We examined Ranavirus transmission between juvenile L. sylvaticus in two settings: first, we measured transmission from a Ranavirus-infected frog to a co-housed susceptible frog via cohabitation over a range of exposure periods. Second, we measured indirect transmission to susceptible frogs from a contaminated environment after a range of waiting times (i.e., from when the infected frog was removed to when the susceptible frog was exposed to the environment). We present evidence that juvenile frogs directly transmitted Ranavirus to susceptible frogs in all exposure periods (99.2% infected), with as little as 1 h of co-housing resulting in 95.8% of susceptibles infected. Indirectly, 96.8% of susceptible frogs became infected after as long as 48 h waiting times. Neither exposure period nor wait times influenced the probability of infection, because these probabilities are already high. In our linear regression models, susceptible frog viral load was significantly correlated with exposure period and cohort for the cohabitation experiment, while wait time was significantly correlated with viral load for the sequential habitation experiment. Collectively, our results suggest that Ranavirus transmission readily occurs between recently metamorphosed wood frogs and via terrestrial environmental exposure.
Collapse
Affiliation(s)
- Maya R Rayle
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA.
| | - Jesse L Brunner
- School of Biological Sciences, Washington State University, Pullman, WA, 99163, USA
| | - Nicole C Dahrouge
- Department of Natural Resources and the Environment, University of Connecticut, Storrs, CT, 06269, USA
| | - Erin L Keller
- School of Biological Sciences, Washington State University, Pullman, WA, 99163, USA
| | - Tracy A G Rittenhouse
- Department of Natural Resources and the Environment, University of Connecticut, Storrs, CT, 06269, USA
| |
Collapse
|
24
|
Soleimani M, Jalilvand A. Spatial analysis of COVID-19 incidence and mortality rates in northwest iran for future epidemic preparedness. Sci Rep 2025; 15:7450. [PMID: 40032988 DOI: 10.1038/s41598-025-91246-y] [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: 10/31/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
The COVID-19 pandemic has underscored the critical need for effective public health strategies to combat infectious diseases. This study examines the epidemiological characteristics and spatial distribution of COVID-19 incidence and mortality in Zanjan Province, northwest Iran, to inform future epidemic preparedness. Using data from 39,739 hospitalized COVID-19 cases recorded between February 2020 and September 2021, sourced from the Medical Care Monitoring Center, we conducted descriptive and geospatial analyses. Demographic, clinical, and spatial variables were analyzed using logistic regression and advanced spatial techniques, including Kernel Density Estimation and Local Moran's I, to identify risk factors and disease hotspots. Results revealed that women accounted for 52% of cases, with higher incidence rates, while men exhibited higher mortality rates (7.86% vs. 7.80%). Urban areas, particularly the provincial capital, were identified as hotspots, with the highest patient density (20,384 cases per 10 km²). Comorbidities such as HIV/AIDS (OR: 4.85), chronic liver disease (OR: 3.6), chronic blood diseases (OR: 2.8), and cancer (OR: 2.5) significantly increased mortality risk, with ventilator use showing the highest odds ratio for death (OR = 91). Vaccination significantly reduced mortality, with fully vaccinated individuals experiencing a 6.3% mortality rate compared to 8.1% in unvaccinated individuals. Spatial analysis highlighted population density and mobility as key drivers of disease spread. These findings emphasize the importance of integrating spatial and epidemiological data to enhance pandemic preparedness. Targeted interventions in urban hotspots, early detection systems, and prioritizing vaccination for high-risk populations are critical for mitigating future outbreaks. This study provides a foundation for evidence-based public health strategies to strengthen global epidemic response and improve preparedness for future health crises.
Collapse
Affiliation(s)
- Mohsen Soleimani
- Assistant Professor of Medical Informatics, Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ahmad Jalilvand
- Associate Professor of Pathology, Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
25
|
Al-Qadami G, Raposo A, Chien CC, Ma C, Priebe I, Hor M, Fung K. Intestinal organoid coculture systems: current approaches, challenges, and future directions. Am J Physiol Gastrointest Liver Physiol 2025; 328:G252-G276. [PMID: 39716040 DOI: 10.1152/ajpgi.00203.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
The intestinal microenvironment represents a complex and dynamic ecosystem, comprising a diverse range of epithelial and nonepithelial cells, a protective mucus layer, and a diverse community of gut microbiota. Understanding the intricate interplay between these components is essential for uncovering the mechanisms underlying intestinal health and disease. The development of intestinal organoids, three-dimensional (3-D) mini-intestines that closely mimic the architecture, cellular diversity, and functionality of the intestine, offers a powerful platform for investigating different aspects of intestinal physiology and pathology. However, current intestinal organoid models, mainly adult stem cell-derived organoids, lack the nonepithelial and microbial components of the intestinal microenvironment. As such, several coculture systems have been developed to coculture intestinal organoids with other intestinal elements including microbes (bacteria and viruses) and immune, stromal, and neural cells. These coculture models allow researchers to recreate the complex intestinal environment and study the intricate cross talk between different components of the intestinal ecosystem under healthy and pathological conditions. Currently, there are several approaches and methodologies to establish intestinal organoid cocultures, and each approach has its own strengths and limitations. This review discusses the existing methods for coculturing intestinal organoids with different intestinal elements, focusing on the methodological approaches, strengths and limitations, and future directions.
Collapse
Affiliation(s)
| | - Anita Raposo
- Health and Biosecurity, CSIRO, Sydney, New South Wales, Australia
| | - Chia-Chi Chien
- Australian Animal Health Laboratory, Australian Centre for Disease Preparedness, CSIRO, Geelong, Victoria, Australia
| | - Chenkai Ma
- Health and Biosecurity, CSIRO, Sydney, New South Wales, Australia
| | - Ilka Priebe
- Health and Biosecurity, CSIRO, Adelaide, South Australia, Australia
| | - Maryam Hor
- Health and Biosecurity, CSIRO, Adelaide, South Australia, Australia
| | - Kim Fung
- Health and Biosecurity, CSIRO, Sydney, New South Wales, Australia
| |
Collapse
|
26
|
Debsikréo N, Dehainsala M, Debsikréo O, Leye N, LO G, Dia A, Flore MNB, Diaw NA, Diouf ND, Otchere ID, Toyé RM, Chemin I, Moussa AM, Toure-Kane NC, Lunel-Fabiani F. Prevalence and molecular characterization of hepatitis delta virus infection among hepatitis B virus surface antigen positive students and pregnant women in N'djamena, Chad. IJID REGIONS 2025; 14:100560. [PMID: 39895833 PMCID: PMC11786080 DOI: 10.1016/j.ijregi.2024.100560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 02/02/2025]
Abstract
Objectives This study sought to determine the prevalence of hepatitis B virus (HBV)-hepatitis D virus (HDV) co-infection and to characterize isolates of both viruses in a Chadian population of HBV surface antigen (HBsAg)-positive pregnant women and students. Methods This was a cross-sectional retrospective study using archived samples from pregnant women and students in N'djamena who had been systematically screened for HBsAg between April and August 2021. HBsAg-positive samples were tested for the presence of HDV antibodies (Ab) and were screened for the presence of both HBV and HDV (in anti-HDV Ab-positive samples) viral load estimations. Genome sequencing of the viruses was used for both genotyping and phylogenetic analysis. Results A total of 94 participants were included in this study. The mean age was 24 ± 4.89 years (range: 18-42 years). Anti-HDV Ab were found in 9.57% (9/94) of the participants. The prevalence of anti-HDV Ab positivity among students (6.45% [4/62]) was lower than the 15.63% (5/32) observed among pregnant women. HDV-RNA was detected in 7/9 (77.77%) confirmed anti-HDV-positive participants. Most HDV-RN-positive participants had very low HBV DNA viral loads. All HBV sequences belonged to genotype E and all HDV sequences to genotype 1. Conclusions Hepatitis D is a potential public health challenge in Chad, which requires active surveillance and public education in the country for proper control. This surveillance should be supported with mass immunization against HBV.
Collapse
Affiliation(s)
- Nalda Debsikréo
- Cheikh Anta Diop University, Dakar, Senegal
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
- University of N'Djamena, N'Djamena, Chad
| | | | - Odan Debsikréo
- University of Félix Houphouët Boigny Abidjan, Abidjan, Côte d'ivore
| | - Nafissatou Leye
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | - Gora LO
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | - Aminata Dia
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | | | - Ndeye Aminata Diaw
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | - Ndeye Dieynaba Diouf
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | - Isaac Darko Otchere
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Fajara, the Gambia
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Rayana Maryse Toyé
- Centre de Recherche en Cancérologie de Lyon INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
| | - Isabelle Chemin
- Centre de Recherche en Cancérologie de Lyon INSERM U1052, CNRS UMR5286, Université de Lyon, Lyon, France
| | - Ali Mahamat Moussa
- University of N'Djamena, N'Djamena, Chad
- Centre Hospitalier Universitaire la Référence, N'Djamena, Chad
| | - Ndèye Coumba Toure-Kane
- Institut de Recherche en Santé, de Surveillance Épidémiologique et de Formation, Dakar, Sénégal
| | - Françoise Lunel-Fabiani
- Centre Hospitalier Universitaire Angers, BAT IBS-4 rue Larrey-49000 ANGERS, Laboratoire HIFIH, UFR Santé département Médecine, SFR 4208-UPRES EA3859, Université d'Angers, Angers Cedex, France
| |
Collapse
|
27
|
Park S, Lee YW, Choi S, Jo H, Kim N, Cho S, Lee E, Choi EB, Park I, Jeon Y, Noh H, Seok SH, Oh SH, Choi YK, Kwon HK, Seo JY, Nam KT, Park JW, Choi KS, Lee HY, Yun JW, Seong JK. Post-COVID metabolic enzyme alterations in K18-hACE2 mice exacerbate alcohol-induced liver injury through transcriptional regulation. Free Radic Biol Med 2025; 229:1-12. [PMID: 39798903 DOI: 10.1016/j.freeradbiomed.2025.01.015] [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: 11/02/2024] [Revised: 12/22/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a significant threat to global public health. Despite reports of liver injury during viral disease, the occurrence and detailed mechanisms underlying the development of secondary exogenous liver injury, particularly in relation to changes in metabolic enzymes, remain to be fully elucidated. Therefore, this study was aimed to investigate the mechanisms underlying SARS-CoV-2-induced molecular alterations in hepatic metabolism and the consequent secondary liver injury resulting from alcohol exposure. We investigated the potential effects of SARS-CoV-2 infection on alcohol-induced liver injury in Keratin 18 promoter-human angiotensin converting enzyme 2 (K18-hACE2) transgenic mice. Mice were intranasally infected with 1 × 102 PFU of SARS-CoV-2. Following a 14 d recovery period from infection, the recovered mice were orally administered alcohol at 6 g/kg. Prior SARS-CoV-2 infection aggravated alcohol-induced liver injury based on increased alanine aminotransferase levels and cytoplasmic vacuolation. Interestingly, infected mice exhibited lower blood alcohol levels and higher levels of acetaldehyde, a toxic alcohol metabolite, compared to uninfected mice after the same period of alcohol consumption. Along with alterations of several metabolic process-related terms identified through RNA sequencing, notably, upregulation of cytochrome P450 2E1 (CYP2E1) and CYP1A2 was observed in infected mice compared to control value prior to alcohol exposure, with no significant impact of SARS-CoV-2 on intestinal damage. Tumor necrosis factor-alpha persistently showed upregulated expression in the infected mice; it also enhanced aryl hydrocarbon receptor and Sp1 expressions and their binding activity to Cyp1a2 and Cyp2e1 promoters, respectively, in hepatocytes, promoting the upregulation of their transcription. Our findings suggest that SARS-CoV-2 infection exacerbates alcohol-induced liver injury through the transcriptional activation of Cyp1a2 and Cyp2e1, providing valuable insights for the development of clinical recommendations on long COVID.
Collapse
Affiliation(s)
- SiYeong Park
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 23488, Republic of Korea
| | - Seunghoon Choi
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - NaHyun Kim
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eunji Lee
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun-Bin Choi
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Inyoung Park
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jeon
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Hyuk Seok
- Laboratory of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun Oh
- Laboratory of Histology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun-Young Seo
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun Won Park
- Laboratory of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, BK21 PLUS Program for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 23488, Republic of Korea.
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea; Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
28
|
Cordie A, Mohamed R, Hatem A, Essam M, Shaltout SW, El Garhy N, Al Sehemy L, Sherif M, Awad RA, Abdelraouf MI, Al-Sharif AM, Ramadan A, Hassany S, El Khateeb E, Sayed AM, Masoud ZWZ, Hamza RS, Tamim HH, Eldin Hamdy MS, Esmat G, Karam-Allah Ramadan H. Retention in care among people living with human immunodeficiency virus (HIV) in a low-resource setting. Int J STD AIDS 2025; 36:275-282. [PMID: 39655701 DOI: 10.1177/09564624241306152] [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: 03/05/2025]
Abstract
BACKGROUND This study aimed to evaluate the retention in care among a cohort of Egyptian people living with HIV (PLWHIV). METHOD The study was conducted on PLWHIV attending Kasr Alainy HIV and Viral Hepatitis Centre, Cairo, Egypt, from January 1, 2019, to March 31, 2023. PLWHIV were considered not retained in care if there was no documented clinical visit or HIV viral load (VL) or CD4 count test for more than 6 months from their last recorded visit or test. Multivariable logistic regression analysis was used to test factors associated with retention in care. RESULTS After excluding those who died and were referred, 369 PLWHIV were included in the analysis, and retention in care was observed in 325 (88%). The majority were males (81.8%) with a median age of 34 [29-41] years. Undetectable VL (OR: 3.555; 95% CI: 1.49-8.47), hepatitis B vaccination (OR: 2.835; 95% CI: 1.07-7.48), CD4 test availability (OR: 2.604; 95% CI: 1.02-6.64), receiving dolutegravir based antiretroviral therapy (OR: 2.429; 95% CI: 1.06-5.537), and longer duration of know HIV infection (OR: 1.025; 95% CI: 1.01- 1.04) were correlated with retention in care. Surprisingly, higher education levels were negatively correlated with retention in care (OR: 0.195, 95%: CI: 0.071-0.533), suggesting the need for further research to explore this relationship. CONCLUSION These results are invaluable for developing targeted interventions and informing health policies to improve retention in HIV care in Egypt. Enhancing access to VL and CD4 testing, promoting VL suppression, and focusing on specific groups at risk of dropping out of care are essential strategies.
Collapse
Affiliation(s)
- Ahmed Cordie
- Endemic Medicine Department, Cairo University, Cairo, Egypt
- Kasr Al-Aini HIV and Viral Hepatitis Fighting Group, Cairo University Hospitals, Cairo University, Cairo, Egypt
| | - Rahma Mohamed
- Endemic Medicine Department, Cairo University, Cairo, Egypt
- Kasr Al-Aini HIV and Viral Hepatitis Fighting Group, Cairo University Hospitals, Cairo University, Cairo, Egypt
| | - Ammar Hatem
- Endemic Medicine Department, Cairo University, Cairo, Egypt
| | - Mahmoud Essam
- Endemic Medicine Department, Cairo University, Cairo, Egypt
| | | | | | | | - Mirella Sherif
- Endemic Medicine Department, Cairo University, Cairo, Egypt
| | | | | | | | - Ahmed Ramadan
- Department of Chemical & Biotechnology Engineering, Sherbrooke, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sahar Hassany
- Department of Tropical Medicine and Gastroenterology, Assiut University, Assiut, Egypt
| | - Engy El Khateeb
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | - Amal M Sayed
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | | | - Rania Soliman Hamza
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | - Hend Hamed Tamim
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | | | - Gamal Esmat
- Endemic Medicine Department, Cairo University, Cairo, Egypt
- Badr University in Cairo, Research Center, Badr City, Cairo, Egypt
| | | |
Collapse
|
29
|
Sun M, Lu Z, Chen WM, Lv S, Fu N, Yang Y, Wang Y, Miao M, Wu SY, Zhang J. N-acetylcysteine therapy reduces major adverse cardiovascular events in patients with type 2 diabetes mellitus. Atherosclerosis 2025; 402:119117. [PMID: 39903949 DOI: 10.1016/j.atherosclerosis.2025.119117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/20/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Effective preventive strategies for major adverse cardiovascular events (MACE) in T2DM patients are limited. Recent studies have explored the cardiovascular benefits of N-Acetylcysteine (NAC), an antioxidant with endothelial protective properties. This study investigates the long-term effects of NAC on MACE risk in T2DM patients, focusing on its potential as an adjunctive therapy. METHODS This population-based cohort study used data from Taiwan's National Health Insurance Research Database (NHIRD) and included 46,718 T2DM patients diagnosed between 2008 and 2018, with follow-up until December 31, 2021. Propensity score matching (PSM) ensured balanced comparisons between NAC users and non-users. Cox regression and time-dependent Cox hazards models assessed MACE risk, adjusting for multiple covariates. RESULTS In the matched cohort of 23,359 NAC users and 23,359 non-users, NAC users had a significantly lower incidence of MACE (41.74 % vs. 46.87 %, P < .0001). Adjusted Hazard Ratios (aHRs) indicated a consistent protective effect of NAC against overall MACE (aHR: 0.84; 95 % CI: 0.81-0.86, P < .0001). Higher cumulative defined daily doses (cDDD) of NAC correlated with reduced MACE risk, with the highest quartile (Q4) showing an aHR of 0.61 (95 % CI: 0.58-0.64, P < .0001). CONCLUSION This study underscores the significant reduction in MACE risk among T2DM patients with long-term NAC therapy. Notably, the findings emphasize NAC's dose-dependent effectiveness in diminishing MACE incidence, indicating its potential as a valuable adjunctive therapy for managing cardiovascular risk in T2DM patients.
Collapse
Affiliation(s)
- Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Zhongyuan Lu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Shuang Lv
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Ningning Fu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yitian Yang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yangyang Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Mengrong Miao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan; Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan; Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Management, College of Management, Fo Guang University, Yilan, Taiwan.
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
30
|
Jensen AL, Litorell J, Grip J, Dahlberg M, Joelsson-Alm E, Jonmarker S. A descriptive, retrospective single-centre study of air-leak syndrome in intensive care unit patients with COVID-19. Acta Anaesthesiol Scand 2025; 69:e14582. [PMID: 39936659 DOI: 10.1111/aas.14582] [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: 07/24/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Acute respiratory failure is the predominant presentation of intensive care unit (ICU) patients with COVID-19, and lung protective strategies are recommended to mitigate additional respiratory complications such as air-leak syndrome. The aim of this study is to investigate the prevalence, type, and timing of air-leak syndrome with regards to associated factors and patient outcome in patients with COVID-19 in ICUs at a large Swedish emergency hospital. METHODS This retrospective study included all adult patients admitted to an ICU for COVID-19-related respiratory failure at Södersjukhuset between March 6, 2020, and June 6, 2021. Primary outcomes were proportion of patients diagnosed with air-leak syndrome and its different types of manifestations, and timing of diagnoses in relation to ICU admission and initiation of invasive ventilation. Secondary outcomes included the highest level of respiratory support prior to the diagnosis of air-leak syndrome, patient characteristics and treatment variables associated with air-leak syndrome, and 90-day mortality for patients with air-leak syndrome compared to those without. RESULTS Out of a total of 669 patients, 81 (12%) were diagnosed with air-leak syndrome. Air-leak syndrome manifested as pneumomediastinum (PMD) (n = 58, 72%), pneumothorax (PTX) (n = 43, 53%), subcutaneous emphysema (SCE) (n = 28, 35%) and pneumatocele (PC) (n = 4, 4.9%). Air-leak syndrome was diagnosed at a median of 14 days (IQR 6-22) after ICU admission and 12 days (IQR 6-19) following the initiation of invasive ventilation. The highest respiratory support prior to diagnosis was invasive ventilation (IV) in 64 patients (79%), non-invasive ventilation in two patients (2.5%), and low- or high-flow oxygen in 15 patients (19%). Multiple logistic regression showed that pulmonary disease at baseline (OR 1.87, 95% CI 1.07-3.25), a lower body mass index (OR 0.95, 95% CI 0.9-0.99), admission later compared with earlier in the pandemic (OR 3.89, 95% CI 2.14-7.08), and IV (OR 3.92, 95% CI 2.07-7.44) were associated with an increased risk of air-leak syndrome. Compared with patients not diagnosed with air-leak syndrome, patients with air-leaks had a higher mortality at 90 days after ICU admission, 46% versus 26% (p <.001). However, the mortality rate differed with different air-leak manifestations, 47% for PMD, 47% for PTX, 50% for the combination of both PMD and PTX and 0% in patients with only SCE and/or PC, respectively. CONCLUSION In 669 ICU patients with COVID-19, 12% had one or more manifestations of air-leak syndrome. Notably, PMD, rather than PTX, was the most common manifestation, suggesting a potentially distinctive feature of COVID-19-related air-leak syndrome. Further research is needed to determine whether COVID-19 involves different pathophysiological or iatrogenic mechanisms compared with other critical respiratory conditions. REGISTRATION OF CLINICAL TRIAL Clinicaltrials.gov, identifying number, NCT05877443. EDITORIAL COMMENT This single-centre cohort study of air leakage into soft tissue in ventilated COVID cases presents findings for associated factors and clinical manifestations, including with different COVID-19 periods and treatments.
Collapse
Affiliation(s)
- Alice Löwing Jensen
- Department of Anaesthesia and Intensive Care, Södersjukhuset, Stockholm, Sweden
| | - Jacob Litorell
- Department of Anaesthesia and Intensive Care, Södersjukhuset, Stockholm, Sweden
| | - Jonathan Grip
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Dahlberg
- Department of Surgery, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Eva Joelsson-Alm
- Department of Anaesthesia and Intensive Care, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Jonmarker
- Department of Anaesthesia and Intensive Care, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Gondelaud F, Leval J, Arora L, Walimbe A, Bignon C, Ptchelkine D, Brocca S, Mukhopadyay S, Longhi S. Unraveling the molecular grammar and the structural transitions underlying the fibrillation of a viral fibrillogenic domain. Protein Sci 2025; 34:e70068. [PMID: 39985377 PMCID: PMC11845978 DOI: 10.1002/pro.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/24/2025]
Abstract
Hendra virus (HeV) is a biosafety level 4 human pathogen belonging to the Henipavirus genus within the Paramyxoviridae family. In HeV, the phosphoprotein-encoding gene also drives the synthesis of the V and W proteins that are two major players in the host innate immune response evasion. These three proteins share a common intrinsically disordered N-terminal domain (NTD) and have distinct C-terminal domains. We recently reported the ability of a short region (i.e., PNT3), located within the shared NTD, to form fibrils. We subsequently identified a PNT3 motif (EYYY) critically involved in fibrillation and deciphered the contribution of each tyrosine to the process. Herein, we combined mutational studies with various biochemical and biophysical approaches to further investigate the molecular mechanisms underlying PNT3 fibrillation. The results show that (i) lysine residues play a critical role in driving fibrillation, (ii) hydrophobic residues affect the nucleation step, and (iii) charge distribution strongly affects the fibrillation propensities. Vibrational Raman spectroscopy data further validated the role of lysine residues in promoting fibrillation and enabled documenting the formation of cross-β amyloid structures. Altogether, these results illuminate the molecular mechanisms involved in fibril formation and pave the way towards the rational design of inhibitors.
Collapse
Affiliation(s)
- Frank Gondelaud
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB)UMR 7257, Aix‐Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Julien Leval
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB)UMR 7257, Aix‐Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Lisha Arora
- Centre for Protein Science, Design and Engineering, Department of Chemical Sciences, and Department of Biological SciencesIndian Institute of Science Education and Research (IISER) MohaliMohaliPunjabIndia
| | - Anuja Walimbe
- Centre for Protein Science, Design and Engineering, Department of Chemical Sciences, and Department of Biological SciencesIndian Institute of Science Education and Research (IISER) MohaliMohaliPunjabIndia
| | - Christophe Bignon
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB)UMR 7257, Aix‐Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Denis Ptchelkine
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB)UMR 7257, Aix‐Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Stefania Brocca
- Department of Biotechnology and BiosciencesUniversity of Milano‐BicoccaMilanItaly
| | - Samrat Mukhopadyay
- Centre for Protein Science, Design and Engineering, Department of Chemical Sciences, and Department of Biological SciencesIndian Institute of Science Education and Research (IISER) MohaliMohaliPunjabIndia
| | - Sonia Longhi
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB)UMR 7257, Aix‐Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| |
Collapse
|
32
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Lakshmanappa YS, Shang P, Renu S, Dhakal S, Hogshead B, Xiao Y, Wang T, Fang Y, Renukaradhya GJ. Concurrent but consecutive vaccination of modified live PRRSV-1 and PRRSV-2 provides better protection in nursery pigs. Vet Microbiol 2025; 302:110391. [PMID: 39854919 DOI: 10.1016/j.vetmic.2025.110391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) virus is a severe threat to the global swine industry. Modified live virus vaccines (MLVs) for two PRRSV species (PRRSV-1 and PRRSV-2) are the most widely used approach to control PRRSV-caused diseases. For swine herds influenced by PRRSV-1 and PRRSV-2, how to rationalize MLV immunization strategies for robust and cross-protective immune responses has been a long-lasting need. In this study, we found that the replication of PRRSV-1 is strongly suppressed by co-infection with PRRSV-2 in vitro, especially under concurrent co-infection conditions. We compared the adaptive immune responses between consecutive and concurrent vaccination methods in nursery pigs, vaccinated either 3 days apart (PRRSV-1 MLV followed by PRRSV-2 MLV, consecutive) or together on the same day (concurrent). PRRSV-1 RNAs were mainly detectable in the sera of consecutively vaccinated pigs. In contrast, PRRSV-2 RNAs in sera were not changed in both vaccination strategies. After the homologous PRRSV-1 or PRRSV-2 challenge, we found that consecutive vaccination slightly improved PRRSV-1 viremia clearance and did not attenuate the PRRSV-2 viremia clearance. Both vaccination strategies induced comparable T-helper cell responses against PRRSV-1 and PRRSV-2 in peripheral blood before and after the challenge. Interestingly, consecutive vaccination induced significantly higher PRRSV-1-specific post-challenge T-helper and cytotoxic T cells responses in the tracheobronchial lymph nodes than concurrent vaccination. Furthermore, consecutive vaccination significantly improved neutralizing antibody responses against PRRSV-1 and PRRSV-2 in comparison with concurrent vaccination. In conclusion, consecutive vaccination appears to be better for viral clearance and induction of adaptive immune response, and our study provides a preliminary rationale to optimize PRRS MLV immunization strategy for better dual protection.
Collapse
Affiliation(s)
- Yashavanth Shaan Lakshmanappa
- Center for Food Animal Health (CFAH), Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, United States
| | - Pengcheng Shang
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Sankar Renu
- Center for Food Animal Health (CFAH), Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, United States
| | - Santosh Dhakal
- Center for Food Animal Health (CFAH), Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, United States
| | - Bradley Hogshead
- Center for Food Animal Health (CFAH), Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, United States
| | - Yihong Xiao
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Tao Wang
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Ying Fang
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States; Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States.
| | - Gourapura J Renukaradhya
- Center for Food Animal Health (CFAH), Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, United States.
| |
Collapse
|
34
|
Khatiwada S, Ngunjiri J, Boley PA, Yadav KK, Ghorbani A, Abundo M, Lee CM, Poelstra JW, Lee CW, Gharaibeh S, Rajashekara G, Kenney SP. Age-based host response to Turkey arthritis reovirus in commercial Turkeys in the presence of maternally derived antibodies. BMC Vet Res 2025; 21:96. [PMID: 39994714 PMCID: PMC11852562 DOI: 10.1186/s12917-025-04525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Turkey arthritis reovirus (TARV) causes arthritic lameness in market-age turkeys. Since 2011, highly pathogenic TARV strains have caused significant economic losses in the turkey industry due to increased culling, reduced market weights, and decreased carcass quality, necessitating more effective control measures. Autogenous vaccine prevention strategies have been inefficacious partly due to a limited understanding of age-related susceptibility of turkeys to TARV. This study investigated age-related host and gut microbiota responses to TARV infection in commercial turkeys derived from vaccinated breeder hens. Poults with known maternally derived antibody titers were orally challenged with TARV O'Neil strain at 1-, 3-, and 7- weeks of age (WOA) and monitored for cloacal virus shedding, gastrocnemius tendon viral tropism, tendon inflammation, weight gain, and changes in gut microbiota. RESULTS A transient TARV-induced weight gain suppression was evident in poults infected at 1- and 3- WOA during the first 3 weeks post-infection. Age-dependent variations in cloacal viral shedding, virus isolation from tendons, and tendon inflammation severity were also observed. There was significant dissimilarity in ileal and cecal bacterial communities between mock and infected groups, but the effect of age of infection was unclear. CONCLUSIONS Age dependent host response was observed to TARV based on cloacal virus shedding, weight gain suppression and viral tendon tropism. Our study also indicates that maternally derived antibodies appeared insufficient to prevent virus translocation to the tendons and subsequent pathological changes. This study lays the groundwork for future investigations of better vaccines/vaccination strategies and alternative preventive measures. IMPORTANCE Turkey arthritis reovirus (TARV) causes lameness due to arthritis and tenosynovitis, commonly in market-age turkeys, resulting in significant economic losses. As a control strategy, the turkey industry used autogenous vaccines, prepared from field TARV isolates in breeder hens, to protect the poults in the early stage of life through maternally derived antibodies (MDAs). This study establishes the level of protection provided by MDAs in young poults with age-based responses to TARV O'Neil reovirus strain. Additionally, this study reveals the dynamics of gut dysbiosis in infected poults at different timepoints, paving the way to ground-breaking investigations into gut microbiome modulation interventions that could potentially improve vaccine efficacy and reduce virus transmission and disease severity.
Collapse
Affiliation(s)
- Saroj Khatiwada
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| | | | - Patricia A Boley
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| | - Kush K Yadav
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Amir Ghorbani
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Carolyn M Lee
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Jelmer W Poelstra
- Molecular and Cellular Imaging Center (MCIC), College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
| | - Chang-Won Lee
- Southeast Poultry Research Laboratory, National Poultry Research Center, USDA Agricultural Research Service, United States Department of Agriculture, Athens, GA, USA
| | - Saad Gharaibeh
- Department of Pathology and Animal Health, Jordan University of Science and Technology, Irbid, Jordan
| | - Gireesh Rajashekara
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA
| | - Scott P Kenney
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agriculture and Environmental Sciences, The Ohio State University, Wooster, OH, USA.
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
35
|
Cea-Callejo P, Trenado C, El Mansouri E, Gomez-Lucia E, Doménech A, Biarnés M, Cuenca JM, Sánchez-Llatas CJ, Madrid R, Benítez L. Point-of-Care Diagnostic Test for Rapid Detection of Infectious Laryngotracheitis Virus by Loop-Mediated Isothermal Amplification and Nanoprobes. Int J Mol Sci 2025; 26:1971. [PMID: 40076597 PMCID: PMC11899946 DOI: 10.3390/ijms26051971] [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: 01/14/2025] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Infectious laryngotracheitis virus (ILTV), a DNA virus classified as Gallid alphaherpesvirus 1, causes a highly contagious respiratory disease in chickens, leading to significant economic losses and health risks for the poultry industry. The rapid detection of ILTV is essential to control its spread and prevent outbreaks. Traditional diagnostic methods like PCR are costly, require specialized personnel, and delay response efforts. To address this, we developed a point-of-care diagnostic test combining loop-mediated isothermal amplification (LAMP) with DNA nanoprobes on respiratory swabs. LAMP targets the ILTV-glycoprotein E (gE) gene, enabling rapid nucleic acid amplification at 65 °C without extraction, making it suitable for on-site detection. DNA nanoprobes provide a colorimetric readout visible to the naked eye. Gold nanoparticles drive this readout, as their red color, based on localized surface plasmon resonance, persists in the presence of ILTV DNA through DNA-DNA hybridization, ensuring reliable detection. The assay achieved 100% sensitivity and specificity for ILTV-gE, with a detection limit of 200 copies per reaction, allowing for the early identification of infections. The results are available within 45 min, enabling prompt measures to control ILTV spread. Cost-effective and user-friendly, this method enhances disease management and biosecurity in poultry farms.
Collapse
Affiliation(s)
- Pablo Cea-Callejo
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
- Research Group of “Animal Viruses”, Complutense University of Madrid, 28040 Madrid, Spain; (E.G.-L.); (A.D.)
| | - Claudia Trenado
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
| | - Elías El Mansouri
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
| | - Esperanza Gomez-Lucia
- Research Group of “Animal Viruses”, Complutense University of Madrid, 28040 Madrid, Spain; (E.G.-L.); (A.D.)
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Ana Doménech
- Research Group of “Animal Viruses”, Complutense University of Madrid, 28040 Madrid, Spain; (E.G.-L.); (A.D.)
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Mar Biarnés
- Centro de Sanidad Avícola de Cataluña y Aragón (CESAC), 43206 Reus, Spain;
| | - J. Marco Cuenca
- Department of Physical Chemistry, School of Chemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain;
| | - Christian J. Sánchez-Llatas
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
| | - Ricardo Madrid
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
- Research Group of “Animal Viruses”, Complutense University of Madrid, 28040 Madrid, Spain; (E.G.-L.); (A.D.)
| | - Laura Benítez
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (P.C.-C.); (C.T.); (E.E.M.); (C.J.S.-L.)
- Research Group of “Animal Viruses”, Complutense University of Madrid, 28040 Madrid, Spain; (E.G.-L.); (A.D.)
| |
Collapse
|
36
|
Himmler GE, Mladinich MC, Conde JN, Gorbunova EE, Lindner MR, Kim HK, Mackow ER. Passage-attenuated Powassan virus LI9P protects mice from lethal LI9 challenge and links envelope residue D308 to neurovirulence. mBio 2025:e0006525. [PMID: 39998203 DOI: 10.1128/mbio.00065-25] [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: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Powassan virus (POWV) is an emergent tick-borne flavivirus that causes lethal encephalitic disease and chronic neurologic deficits in surviving patients. POWV-LI9 is a tick-derived isolate that causes neurovirulent disease and age-dependent lethality in mice. Serial passage of VeroE6 cells infected with LI9 resulted in eight amino acid changes in a POWV strain LI9P. LI9P fails to cause neurological sequelae, or lethality in C57BL/6 mice yet elicits neutralizing POWV antibody responses and protects mice from lethal LI9 challenge. Analysis revealed that LI9, but not LI9P, is present at high levels in the CNS, suggesting that LI9P is restricted from neuroinvasion or CNS replication. LI9 and LI9P are distinguished by a D308N envelope change within a domain associated with cell attachment. We evaluated the roles of Env-Domain III residue changes in LI9 virulence and LI9P attenuation using recombinant POWVs (recPOWVs) generated by reverse genetics. Remarkably, mutating D308N in LI9 completely abolished viral lethality and neuroinvasion in 50-week-old mice, reflecting the avirulent phenotype of LI9P. Analysis of the reciprocal N308D change in LI9P only partially restored neuroinvasion and lethality to the LI9P-N308D mutant, indicating that further LI9P residue changes contribute to LI9P attenuation. Consistent with differences in neuroinvasion, we found that rapid LI9P RNA synthesis and corresponding early IFN induction may contribute to LI9P clearance. Collectively, these findings define D308 as a determinant of POWV neuroinvasion and lethality, suggest potential mechanisms for restricted LI9P CNS entry, and reveal passage-attenuated LI9P as a candidate POWV vaccine platform. IMPORTANCE Powassan virus (POWV) infection causes a 10% lethal encephalitis, resulting in chronic neurological symptoms in half of survivors. POWV is transmitted in as short as 15 min following tick attachment, demonstrating the need for the development of POWV vaccines and therapeutics. Mechanisms of POWV neurovirulence remain to be defined to inform vaccine and therapeutic design. Cell culture passage has successfully been used to generate live-attenuated flavivirus vaccines. Accordingly, we serially passaged POWV LI9-infected VeroE6 cells and isolated an attenuated POWV strain, LI9P, that fails to cause neurologic sequelae or murine lethality. LI9P elicits neutralizing antibody responses, protects mice from a lethal WT POWV challenge, and is a potential POWV vaccine. Analysis of attenuating mutations in LI9P revealed that changing envelope residue D308N alone in LI9 prevents POWV neurovirulence and lethality in immunocompetent mice. Altogether, this study defines viral determinants of POWV pathogenesis and attenuating mutations that inform the development of live-attenuated POWV vaccines.
Collapse
Affiliation(s)
- Grace E Himmler
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Megan C Mladinich
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Department of Biological Sciences, SUNY Old Westbury, Old Westbury, New York, USA
| | - Jonas N Conde
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Elena E Gorbunova
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Marissa R Lindner
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Hwan Keun Kim
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Erich R Mackow
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
37
|
Chen ZY, Zhang J, He PJ, Xiong T, Zhu DY, Zhu WJ, Ni XB, Du LF, Wang Q, Zhang YW, Xia LY, Chen DS, Li LJ, Zhang MZ, Cui XM, Wang TH, Wang J, Wang Z, An TF, Cao WC, Liu XH, Huang EJ, Jia N. Characteristics of viral ovarian tumor domain protease from two emerging orthonairoviruses and identification of Yezo virus human infections in northeastern China as early as 2012. J Virol 2025; 99:e0172724. [PMID: 39745436 PMCID: PMC11852922 DOI: 10.1128/jvi.01727-24] [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: 10/07/2024] [Accepted: 12/05/2024] [Indexed: 02/26/2025] Open
Abstract
Emerging tick-borne orthonairovirus infections pose a growing global concern, with limited understanding of the viral ovarian tumor-like cysteine proteases (vOTUs) encoded by novel orthonairoviruses. These vOTUs, a group of deubiquinylases (DUBs), disrupt the innate immune response. Yezo virus (YEZV), a recently discovered pathogenic orthonairovirus, was first reported in Japan in 2021. In this study, we successfully isolated and identified YEZV and a new orthonairovirus, Jiànchuān tick virus (JCTV), from Ixodes persulcatus and Haemaphysalis montgomeryi ticks, respectively, in China. We found that the vOTU domains encoded by YEZV and JCTV exhibited both DUB and deISGylase activities, though with potentially less broad deISGylation compared to that of Crimean-Congo hemorrhagic fever virus (CCHFV) during natural infection. Phylogenetic analysis of global vOTUs, including 83 new sequences, revealed a high diversity of this domain. Interestingly, retrospective screening of tick-bite patients from 2012 to 2016 in northeastern China traced YEZV infections as far back as 2012, identifying four cases. Additionally, YEZV primarily infected I. persulcatus (31.4%) and Dermacentor nuttalli (10.5%) in northern China, while JCTV exhibited high infection rates in H. montgomeryi (81.3%) in southern China. In summary, our work emphasizes the active surveillance of orthonairovirus infections and the imperative need for the development of vOTU domain-targeted anti-virals, offering potential therapeutic solutions for a broad spectrum of orthonairoviruses.IMPORTANCEThe vOTUs, a group of DUBs, mimic the functions of host DUBs to enhance viral infectivity and may serve as potential drug targets. vOTUs from different orthonairoviruses exhibit distinct preferences toward ubiquitin (Ub) and ubiquitin-like protein interferon stimulated gene 15 (ISG15). In this study, we investigated the deubiquitinase and deISGylase functions of various orthonairoviral vOTUs using both an overexpression system and natural viral infections in vitro. Our findings illustrate that the vOTUs from YEZV and JCTV can cleave both Ub and ISG15 in an overexpression system, but these viruses exhibit potentially narrower deISGylation capacity than CCHFV during natural infection. This suggests that the diversity of vOTUs may have a potential relationship with the pathogenesis.
Collapse
Affiliation(s)
- Zi-Yun Chen
- School of Public Health of Fujian Medical University, Fuzhou, Fujian, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Jie Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Pei-Jun He
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tao Xiong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Dai-Yun Zhu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Wen-Jie Zhu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xue-Bing Ni
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, School of Public Health, University of Hong Kong, Hong Kong, China
| | - Li-Feng Du
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, Shanghai, China
| | - Qian Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Ya-Wei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Luo-Yuan Xia
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Dong-Sheng Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Liang-Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Ming-Zhu Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao Ming Cui
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Hong Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Juan Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, Shanghai, China
| | - Zhenfei Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Feng An
- Department of Toxicology and Health Inspection and Quarantine, School of Public Health, Tianjin Medical University, Tianjin, Tianjin, China
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-Hua Liu
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, Jiangxi, China
| | - En-Jiong Huang
- School of Public Health of Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou International Travel Healthcare Center, Fuzhou, China
| | - Na Jia
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
38
|
Yang QL, Lu XW, Fang ZL, Gao YQ, He YN, Huang Y, Dai Y, Liang MY, Chan CHF, Jiang ZH. The association between Clonorchis sinensis seropositivity and hepatocellular carcinoma in an endemic area: a study in Guangxi, China. BMC Infect Dis 2025; 25:270. [PMID: 40000979 PMCID: PMC11852542 DOI: 10.1186/s12879-025-10675-2] [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: 05/20/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Chronic infection with Clonorchis sinensis (C.sinensis) has been associated swith the development of intrahepatic cholangiocarcinoma (ICC); however, the relationship between C.sinensis and hepatocellular carcinoma (HCC) remains uncertain. METHODS This study examined 120 patients with liver cancer in the clonorchiasis endemic area of Hengzhou, Guangxi, China. The type of cancer, the differentiation grade according to Edmondson Steiner's classification, and the pathological characteristics of HCC were determined through postoperative tissue biopsy. C.sinensis infection was detected by measuring serum specific IgG antibody, and hepatitis B virus (HBV) infection was determined by detecting serum HBsAg and HBV DNA in HCC tissues. The C.sinensis infection rates in control groups were drawn from the local general population based on previous surveys. The association between C.sinensis infection and HCC was analyzed by comparing the differences in C.sinensis infection rates between the two groups. RESULTS Of the patients evaluated, 98 (81.7%) had HCC, 21 (17.5%) had ICC, and 1 (0.8%) had comorbidity of HCC/ICC. Among the HCC patients, 24 (24.5%) were solely infected with HBV, 71 (72.4%) were C. sinensis seropositive, and 3 (3.1%) showed no evidence of infection. C. sinensis seropositive rates in HCC patients are much higher than in general outpatient and non-liver cancer inpatients (χ2 = 141.92, p < 0.001), as well as in the local residents (χ2 = 82.61/21.38, p < 0.001). There were no significant differences in the pathological type, differentiation grade, and lesion composition between the tumor associated with C.sinensis/HBV mono- and co-infection (p > 0.05). Among the patients with C.sinensis-related HCC, 8 (8.2%) were solely C.sinensis seropositive, while 63 (64.3%) were co-infected with HBV. Infection with C. sinensis and HBV has a significant impact on the pathological types of liver cancer (χ2 = 22.86, p < 0.001). CONCLUSIONS These findings indicate that HCC still accounts for the majority of liver cancer in this region. In addition to being most commonly related with HBV infection, HCC may also be related to C. sinensis infection. Co-infection of C. sinensis and HBV may enhance the development of HCC in this area. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Qing-Li Yang
- Guangxi University of Chinese Medicine, Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Nanning, Guangxi, 530200, People's Republic of China
| | - Xi-Wei Lu
- People's Hospital of Hengzhou, Nanning, Guangxi, 530300, People's Republic of China
| | - Zhong-Liao Fang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Guangxi Key Laboratory for Viral Hepatitis Prevention and Control, Nanning 530028, Guangxi, People's Republic of China, Nanning, Guangxi, People's Republic of China
| | - Yu-Qiu Gao
- Guangxi University of Chinese Medicine, Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Nanning, Guangxi, 530200, People's Republic of China
| | - Yi-Ning He
- Guangxi University of Chinese Medicine, Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Nanning, Guangxi, 530200, People's Republic of China
| | - Yan Huang
- Guangxi University of Chinese Medicine, Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Nanning, Guangxi, 530200, People's Republic of China
| | - Yue Dai
- Guangxi University of Chinese Medicine, Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Nanning, Guangxi, 530200, People's Republic of China
| | - Ming-Yong Liang
- Hengzshou Center for Disease Prevention and Control, Nanning, Guangxi, 530300, China
| | - Carlos H F Chan
- Department of Surgery, University of Iowa Health Care, Iowa City, IA 52242, USA.
| | - Zhi-Hua Jiang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Guangxi Key Laboratory for Viral Hepatitis Prevention and Control, Nanning 530028, Guangxi, People's Republic of China, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
39
|
Choe S, Jeon M, Yoon H. Advanced Therapeutic Approaches for Metastatic Ovarian Cancer. Cancers (Basel) 2025; 17:788. [PMID: 40075635 PMCID: PMC11898553 DOI: 10.3390/cancers17050788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death among women, which is one of the most common gynecological cancers worldwide. Although several cytoreductive surgeries and chemotherapies have been attempted to address ovarian cancer, the disease still shows poor prognosis and survival rates due to prevalent metastasis. Peritoneal metastasis is recognized as the primary route of metastatic progression in ovarian cancer. It causes severe symptoms in patients, but it is generally difficult to detect at an early stage. Current anti-cancer therapy is insufficient to completely treat metastatic ovarian cancer due to its high rates of recurrence and resistance. Therefore, developing strategies for treating metastatic ovarian cancer requires a deeper understanding of the tumor microenvironment (TME) and the identification of effective therapeutic targets through precision oncology. Given that various signaling pathways, including TGF-β, NF-κB, and PI3K/AKT/mTOR pathways, influence cancer progression, their activity and significance can vary depending on the cancer type. In ovarian cancer, these pathways are particularly important, as they not only drive tumor progression but also impact the TME, which contributes to the metastatic potential. The TME plays a critical role in driving metastatic features in ovarian cancer through altered immunologic interactions. Recent therapeutic advances have focused on targeting these distinct features to improve treatment outcomes. Deciphering the complex interaction between signaling pathways and immune populations contributing to metastatic ovarian cancer provides an opportunity to enhance anti-cancer efficacy. Hereby, this review highlights the mechanisms of signaling pathways in metastatic ovarian cancer and immunological interactions to understand improved immunotherapy against ovarian cancer.
Collapse
Affiliation(s)
- Soohyun Choe
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Minyeong Jeon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| |
Collapse
|
40
|
Lopes PHR, Pereira NMDO, da Rocha MN, Marinho MM, Guedes JM, Rodrigues THS, Do Vale JPC, Marinho ES, Santiago GMP, dos Santos HS. Chemical Composition and Larvicidal Activity Against Aedes aegypti of the Leaf Essential Oils from Croton blanchetianus. Molecules 2025; 30:1034. [PMID: 40076259 PMCID: PMC11901792 DOI: 10.3390/molecules30051034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
The Aedes aegypti mosquito is the primary vector of dengue, a neglected disease and a serious public health problem in tropical countries. The control of this vector has been carried out using chemical insecticides, which impact human health. Thus, it is essential to develop natural larvicides that are less harmful to the environment. This study investigates the circadian cycle and larvicidal activity of essential oils from Croton blanchetianus against Aedes aegypti. The leaf oils were extracted by hydrodistillation and analyzed by GC-MS and GC-FID. The circadian study revealed variations in the chemical composition of oils extracted at different times of the day. The main constituents were α-pinene, β-phellandrene, eucalyptol, β-caryophyllene, bicyclogermacrene, and spathulenol. The larvicidal activity showed LC50 values at the following different collection times: 55.294 ± 3.209 μg/mL at 08:00 h; 95.485 ± 2.684 μg/mL at 12:00 h; and 64.883 ± 1.780 μg/mL at 17:00 h. Molecular docking simulations indicated that α-pinene, β-phellandrene, eucalyptol, and β-caryophyllene strongly interact with the active site of the sterol carrier protein, suggesting their role in larvicidal activity. These findings reinforce the potential of C. blanchetianus essential oils as an alternative for Aedes aegypti control. The predictive pharmacokinetic tests showed a PAMPA profile associated with high effective cellular permeability and microsomal stability, resulting from the metabolic stability of the derivatives (3) eucalyptol and (6) spathulenol, indicating that these compounds have the highest pharmacokinetic viability and low reactivity with respect to organ toxicity.
Collapse
Affiliation(s)
- Pedro Henrique Ribeiro Lopes
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
| | - Nicaely Maria de Oliveira Pereira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60020-181, CE, Brazil; (N.M.d.O.P.); (G.M.P.S.)
| | - Matheus Nunes da Rocha
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
| | - Marcia Machado Marinho
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
| | - Jesyka Macêdo Guedes
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
| | | | - Jean Parcelli Costa Do Vale
- Center for Exact Sciences and Technology, Vale do Acaraú University, Sobral 62040-370, CE, Brazil; (T.H.S.R.); (J.P.C.D.V.)
| | - Emmanuel Silva Marinho
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
| | - Gilvandete Maria Pinheiro Santiago
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60020-181, CE, Brazil; (N.M.d.O.P.); (G.M.P.S.)
- Department of Pharmacy, Federal University of Ceara, Fortaleza 60430-160, CE, Brazil
| | - Hélcio Silva dos Santos
- Postgraduate Program in Natural Sciences, Ceará State University, Fortaleza 60714-903, CE, Brazil; (P.H.R.L.); (M.N.d.R.); (M.M.M.); (J.M.G.); (E.S.M.)
- Center for Exact Sciences and Technology, Vale do Acaraú University, Sobral 62040-370, CE, Brazil; (T.H.S.R.); (J.P.C.D.V.)
| |
Collapse
|
41
|
Gunchenko M, Shapiro C, Jain S, Doozandeh H. Aplastic anaemia, pernicious anaemia and autoimmune thyroiditis following an episode of EBV-associated hepatitis. BMJ Case Rep 2025; 18:e262950. [PMID: 40000037 DOI: 10.1136/bcr-2024-262950] [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: 02/27/2025] Open
Abstract
Epstein-Barr virus (EBV) affects over 90% of the global population and has been linked to several autoimmune disorders. This report describes a patient with EBV-associated hepatitis who subsequently developed aplastic anaemia, pernicious anaemia and autoimmune thyroiditis. The patient was treated with an immunosuppressive regimen with gradual improvement in his pancytopenia and autoimmune thyroiditis. This report highlights the importance of a comprehensive evaluation and close monitoring of patients presenting with acute or recent EBV infection. Clinicians are urged to recognise autoimmune sequelae, as early intervention can be life-saving.
Collapse
MESH Headings
- Humans
- Male
- Anemia, Pernicious/complications
- Anemia, Pernicious/diagnosis
- Anemia, Pernicious/drug therapy
- Anemia, Aplastic/complications
- Thyroiditis, Autoimmune/complications
- Thyroiditis, Autoimmune/diagnosis
- Epstein-Barr Virus Infections/complications
- Epstein-Barr Virus Infections/diagnosis
- Immunosuppressive Agents/therapeutic use
- Hepatitis, Viral, Human/virology
- Hepatitis, Viral, Human/diagnosis
- Hepatitis, Viral, Human/drug therapy
- Adult
Collapse
Affiliation(s)
- Melissa Gunchenko
- Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Chandler Shapiro
- Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Shivi Jain
- Internal Medicine, Hematology/Oncology, Rush University Medical Center, Chicago, Illinois, USA
| | | |
Collapse
|
42
|
Giri S, Batra L. Memory Cells in Infection and Autoimmunity: Mechanisms, Functions, and Therapeutic Implications. Vaccines (Basel) 2025; 13:205. [PMID: 40006751 PMCID: PMC11860616 DOI: 10.3390/vaccines13020205] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Memory cells are central to the adaptive immune system's ability to remember and respond effectively to previously encountered pathogens. While memory cells provide robust protection against infections, they can also contribute to autoimmunity when regulation fails. Here, we review the roles of memory T and B cells in infection and autoimmunity, focusing on their differentiation, activation, effector functions, and underlying regulatory mechanisms. We elaborate on the precise mechanisms by which memory cells contribute to autoimmune diseases, highlighting insights from current research on how pathogenic memory responses are formed and sustained in autoimmunity. Finally, we explore potential therapeutic strategies aimed at modulating memory cells to prevent or treat autoimmune disorders, including B cell-depleting therapies (e.g., Rituximab), T cell-targeting agents (e.g., Abatacept), and cytokine inhibitors (e.g., IL-17 or IL-23 blockers) that are currently used in diseases such as rheumatoid arthritis, multiple sclerosis, and psoriasis.
Collapse
Affiliation(s)
- Shilpi Giri
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lalit Batra
- Center for Predictive Medicine for Biodefence and Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40222, USA;
| |
Collapse
|
43
|
Zangene S, Morovvati H, Anbara H, Bernabò N. Exposure to Polystyrene Microplastic Differentially Affects the Colon and Liver in Adult Male Mice. ENVIRONMENTAL TOXICOLOGY 2025. [PMID: 39967350 DOI: 10.1002/tox.24486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
Microplastics (MPs) have emerged as novel environmental pollutant. Their ubiquity in natural environments and the global dissemination of plastic particles through food and drink have led to the oral ingestion of these particles by all kinds of living organism. In this investigation, male mice were subjected to exposure to 2 μm virgin PS-MPs for 6 weeks. To accomplish this, 36 adult male NMRI mice were gavaged with PS-MPs at concentrations of 0.01, 0.1, and 1 mg/kg body weight. A control group was also accounted for, which received 0.1 mL of distilled water. The results show that the activity of antioxidant enzymes (superoxide dismutase, catalase, and glutathione peroxidase) decreased, while the level of malondialdehyde increased in colon and liver. Additionally, findings showed that PS-MPs can disrupt the integrity of the intestinal barrier and inhibit the secretion of intestinal mucus in mice, disrupt mucin secretion, and cause changes in the tissue structure of the colon and liver. Further information regarding the toxicity of MPs in a terrestrial organism was obtained through this study, which assist in the evaluation of the potential health hazards that PS-MPs may pose to living organisms.
Collapse
Affiliation(s)
- Somaye Zangene
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Morovvati
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hojat Anbara
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Nicola Bernabò
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| |
Collapse
|
44
|
Girma D, Abita Z, Agegnehu W, Asmelash D, Adugna A, Shifera N, Alie MS, Abebe GF. Predictors of anemia among HIV-infected children in Ethiopia: systematic review and meta-analysis. Sci Rep 2025; 15:5968. [PMID: 39966543 PMCID: PMC11836374 DOI: 10.1038/s41598-025-85285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/01/2025] [Indexed: 02/20/2025] Open
Abstract
Anemia continues to be one of the common complications among HIV-infected children. In Ethiopia, though there is a systematic review and meta-analysis study on anemia among HIV-infected children, it only disclosed the effect Highly Active Antiretroviral Treatment on HIV/AIDS-anemia comorbidity, and yet, the meta effect of other potential factors such as type of ART, presence of opportunistic infection, advanced stage of HIV/AIDS, and cotrimoxazole therapy on HIV/AIDS-anemia comorbidity have not been explored in the previous review. Therefore, this systematic review and meta-analysis aimed to identify the factors associated with anemia among HIV-infected children. Moreover, this study provides an up-to-date pooled estimate of anemia among HIV-infected children in Ethiopia. We systematically searched PubMed, HINARI, Science Direct, Cochrane Library, Google Scholar, and African Journals Online on February 3, 2024, to identify relevant primary research articles. The Briggs Institute (JBI) Checklist was used to check the quality of the original studies. Meta package for proportions (Metapro) was used to estimate the pooled prevalence of anemia among HIV-infected children using the random-effects model. Heterogeneity across studies was checked using the I-square test. Funnel plots visual inspection and Egger's tests were done to detect publication bias. The pooled prevalence of anemia among HIV-infected children in Ethiopia was 23.79% (95% CI 17.28, 31.81). Age < 7 years (OR 3.71, 95% CI 2.58; 5.33), advanced HIV disease (OR 2.78, 95% CI 2.00; 3.87), intestinal parasitic infection (OR 2.28, 95% CI 1.02; 5.09), poor ART treatment adherence (OR 1.96, 95% CI 1.23; 3.10), opportunistic infection (OR 2.81, 95% CI 1.59; 4.95), viral load > 1000 copies/ml (OR 4.29, 95% CI 2.28; 8.09), and zidovudine containing regimen (OR 5.07, 95% CI 2.41; 10.64) were identified as factors associated with a higher risk of anemia. Whereas, cotrimoxazole prophylaxis therapy (OR 0.49, 95% CI 0.35; 0.72) reduces the risk of anemia among HIV-infected children. In Ethiopia, anemia remains a public health concern among children living with HIV. Therefore, regular screening and management of anemia are important for HIV-infected children, particularly for those with advanced HIV disease, opportunistic infection, high viral load, and who are taking zidovudine-containing regimens for better clinical outcomes. Moreover, preventive chemotherapy (deworming) and counseling on infection prevention should be provided for children living with HIV to prevent parasitic infection.
Collapse
Affiliation(s)
- Desalegn Girma
- Department of Midwifery, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia.
| | - Zinie Abita
- Department of Public Health, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| | - Wubetu Agegnehu
- Department of Public Health, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| | - Daniel Asmelash
- Department of Medical Laboratory, College of Health Science, Mizan Tepi University, Mizan-Teferi, Ethiopia
| | - Amanuel Adugna
- Department of Midwifery, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| | - Nigusie Shifera
- Department of Public Health, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| | - Melsew Setegn Alie
- Department of Public Health, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| | - Gossa Fetene Abebe
- Department of Midwifery, College of Health Science, Mizan-Tepi University, Mizan-Teferi, Ethiopia
| |
Collapse
|
45
|
Fretwell EC, Houldsworth A. Oncolytic Virus Therapy in a New Era of Immunotherapy, Enhanced by Combination with Existing Anticancer Therapies: Turn up the Heat! J Cancer 2025; 16:1782-1793. [PMID: 40092697 PMCID: PMC11905400 DOI: 10.7150/jca.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/20/2025] [Indexed: 03/19/2025] Open
Abstract
Oncolytic viral therapy is a promising treatment for cancer, where 'cold' tumour cells can become 'hot' to the host immune system. However, with few FDA approved therapies, development of new strategies for more cancer types has been slow and relatively unsuccessful in recent years, Combination therapy has been successful for other types of cancer treatment, therefore, may be a viable alternative to improve the efficacy of oncolytic viral therapy which may reduce some of the adverse events of currently used monotherapies, oncolytic virus therapy and chemotherapy being mutually complimentary with each other. Combining oncolytic viruses with immune checkpoint inhibitors provides a significant increase in efficacy when viral therapy was combined with the drug ipilimumab. Phase I and II studies concluded that combination with chemotherapies was safe and effective but did not significantly improve on current monotherapies. Recent experiments suggest that a combination of CAR-T and CAR--M cells is a promising therapeutic approach but needs to advance to clinical testing to observe the human response to the therapy. Viral combination with ipilimumab showed the highest potential for a successful treatment and clinical trials should be advanced to phase III to find conclusive supporting evidence. This review aims to identify and evaluate the potential of currently evolving oncolytic viral therapy with recent advances in genetic engineering providing enhanced oncolytic activity in the tumour, and addressing the lack of host immune responses in 'cold' tumours, with an additional role in enhancing conventional treatment efficacy with combination therapies. The potential of oncolytic viruses to 'turn up the heat' of a tumour microenvironment immunogenicity in combination with other anticancer treatments, provides a promising future for new cancer therapies.
Collapse
Affiliation(s)
| | - Annwyne Houldsworth
- University of Exeter Medical School, Faculty of Health and Life Sciences, Exeter, EX2 4TH, UK
| |
Collapse
|
46
|
Chen L, Jiang C, Scholle F, Meo AE, Ohata J, Gorman CB, Ghiladi RA. InP-Based Quantum Dots as Photosensitizers in Photodynamic Antimicrobial Materials. ACS APPLIED BIO MATERIALS 2025; 8:1138-1147. [PMID: 39818708 DOI: 10.1021/acsabm.4c01467] [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: 01/18/2025]
Abstract
Ligand-functionalized InP-based quantum dots (QDs) have been developed as an innovative class of nontoxic photosensitizer suitable for antimicrobial applications, aimed at reducing or preventing pathogen transmission from one host to another via high contact surfaces. A hot injection method followed by functionalization via ligand exchange with 9-anthracene carboxylic acid (ACA) yielded the desired core/shell InP/ZnSe/ZnS QDs. Transmission electron microscopy (TEM) revealed these QDs to be uniform in size (∼3.2 nm), with light absorption across the entire visible spectrum (λmax ∼550 nm). Under light excitation at 550 nm, the generation of singlet oxygen (1O2) was evidenced by its characteristic phosphorescence signal at 1278 nm, indicating successful energy transfer from the QDs to surface-anchored ACA ligands, in accordance with a type II mechanism for a photodynamically generated singlet oxygen. The InP/ZnSe/ZnS core/shell QDs were applied to cellulose via dip coating, and the resultant QDs-loaded material was assessed for antimicrobial photodynamic inactivation (aPDI) of both Gram-positive [methicillin-resistant Staphylococcus aureus (MRSA; ATCC-44), vancomycin-resistant Enterococcus faecium (VRE; ATCC-2320)] and Gram-negative [multidrug-resistant Acinetobacter baumannii (MDRAB; ATCC-1605), NDM-1 positive Klebsiella pneumoniae (KP; ATCC-2146)] bacteria under illumination (400-700 nm; 85 mW/cm2; 90 min). The highest inactivation was observed for MRSA, achieving at least 99.999% inactivation (5 log units). Antiviral photodynamic inactivation on human coronavirus 229E (HCoV-229E) and feline calicivirus (FCV) demonstrated complete viral inactivation (to the detection limit). Cytotoxicity studies showed that the QDs are nontoxic to mammalian cells in the dark. Together, these results confirm the promising potential of ligand-functionalized InP-based QDs to be employed as nontoxic photosensitizers as materials in self-sterilizing surfaces.
Collapse
Affiliation(s)
- Lihan Chen
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Chenyu Jiang
- School of Optical and Electronic Information, Suzhou City University, Suzhou, Jiangsu Province 215104, China
| | - Frank Scholle
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Alissa E Meo
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Jun Ohata
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Christopher B Gorman
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Reza A Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
47
|
Ghafarian S, Samavat B, Lee K, Sheikhghomi S, Cheraghpour K, Shukla D, Djalilian AR, Chodosh J, Soleimani M. Clinical strategies to prevent recurrence of Herpes simplex and Herpes zoster following ocular surgery: A comprehensive review with practical guidelines. Surv Ophthalmol 2025:S0039-6257(25)00029-3. [PMID: 39961450 DOI: 10.1016/j.survophthal.2025.02.002] [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: 10/15/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
Recurrences of herpetic infections following intraocular surgeries pose a threat to optimal surgical outcomes. The high prevalence of herpetic diseases require ophthalmologists to be familiar with the special measures in the surgery of these patients. A thorough preoperative assessment and meticulous postoperative surveillance should be tailored for each patient, depending on the surgery and the risk of virus reactivation. We compile the relevant evidence in the literature and provide a comprehensive review of the preoperative assessment and postoperative diagnostic clues and management of the herpetic infections following different types of intraocular surgeries, including cataract surgery, keratoplasty, corneal crosslinking, glaucoma, and refractive surgeries.
Collapse
Affiliation(s)
- Sadegh Ghafarian
- Department of Ophthalmology, Farabi Eye Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bijan Samavat
- Department of Ophthalmology, Velayat Hospital, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Karen Lee
- Department of Ophthalmology, University of North Carolina, NC, USA
| | - Sima Sheikhghomi
- Department of Ophthalmology, Madani Hospital, Alborz University of Medical Sciences, Jahanshahr, Karaj, Alborz Province, Iran
| | - Kasra Cheraghpour
- Department of Ophthalmology, Farabi Eye Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Deepak Shukla
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico School of Medicine, NM, USA
| | | |
Collapse
|
48
|
Sun Y, Wei Y, Han X, Wang Y, Yin Q, Zhang Y, Yang T, Zhang J, Sun K, Fang F, Zhang S, Yuan K, Li M, Zhao G. Effect of Inoculation Volume on a Mouse Model of Influenza Virus Infected with the Same Viral Load. Vaccines (Basel) 2025; 13:173. [PMID: 40006720 PMCID: PMC11860169 DOI: 10.3390/vaccines13020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/23/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Influenza is a highly contagious respiratory disease that poses significant health and economic burdens. Mice are commonly used as animal models for studying influenza virus pathogenesis and the development of vaccines and drugs. However, the viral volume used for nasal inoculation varies substantially in reported mouse influenza infection models, and the appropriate viral dose is crucial for reproducing experimental results. METHODS Mice were inoculated with mouse lung-adapted strains of influenza virus A/Puerto Rico/8/34 (H1N1) via intranasal administration of 10 μL, 20 μL, and 40 μL at doses of 200 plaque-forming units (PFU) and 2000 PFU. This study investigated the impact of varying viral inoculum volumes on murine outcomes at identical doses and assessed the disparities across diverse dosage levels. RESULTS Regarding weight change trajectories, mortalities, lung tissue viral titers, and pathological manifestations, the group that received the 40 μL inoculation volume within the low-dose infection mice (200 PFU) manifested a statistically significant divergence from those inoculated with both the 10 μL and 20 μL volumes. Within the context of high-dose infections (2000 PFU), groups that received inoculation volumes of 20 μL and 40 μL exhibited marked disparities when compared to those receiving the 10 μL volume. CONCLUSIONS Disparities in inoculation volume, even under uniform infection dosages, engender differential outcomes in pathogenicity. Of particular note, the viral replication efficacy at a 20 μL inoculation volume demonstrates conspicuous fluctuations across diverse infection dose regimens.
Collapse
Affiliation(s)
- Yali Sun
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China; (Y.S.); (T.Y.); (K.S.); (K.Y.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Yuwei Wei
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Yuhang Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Tiantian Yang
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China; (Y.S.); (T.Y.); (K.S.); (K.Y.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Jiejie Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Keyu Sun
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China; (Y.S.); (T.Y.); (K.S.); (K.Y.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Feimin Fang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Shuai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Kai Yuan
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China; (Y.S.); (T.Y.); (K.S.); (K.Y.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (Y.W.); (X.H.); (Y.W.); (Q.Y.); (Y.Z.); (J.Z.); (F.F.); (S.Z.)
| | - Guangyu Zhao
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China; (Y.S.); (T.Y.); (K.S.); (K.Y.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
49
|
Saha T, Masum ZU, Biswas A, Mou MA, Ahmed S, Saha T. Inhaled Dry Powder of Antiviral Agents: A Promising Approach to Treating Respiratory Viral Pathogens. Viruses 2025; 17:252. [PMID: 40007007 PMCID: PMC11860668 DOI: 10.3390/v17020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Inhaled dry powder formulations of antiviral agents represent a novel and potentially transformative approach to managing respiratory viral infections. Traditional antiviral therapies in the form of tablets or capsules often face limitations in terms of therapeutic activity, systemic side effects, and delayed onset of action. Dry powder inhalers (DPIs) provide a targeted delivery system, ensuring the direct administration of antivirals to the infection site, the respiratory tract, which potentially enhance therapeutic efficacy and minimize systemic exposure. This review explores the current state of inhaled dry powder antiviral agents, their advantages over traditional routes, and specific formulations under development. We discuss the benefits of targeted delivery, such as improved drug deposition in the lungs and reduced side effects, alongside considerations related to the formulation preparation. In addition, we summarize the developed (published and marketed) inhaled dry powders of antiviral agents.
Collapse
Affiliation(s)
- Tushar Saha
- Mastaplex Ltd., Centre for Innovation, University of Otago, Dunedin 9016, New Zealand
| | - Zia Uddin Masum
- College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA;
| | - Anik Biswas
- Materials and Nanotechnology, North Dakota State University, Fargo, ND 58105, USA;
| | - Moushumi Afroza Mou
- Department of Biological Science, St. John’s University, Queens, New York, NY 11439, USA;
| | - Sohag Ahmed
- Department of Chemistry, West Virginia University, Morgantown, WV 26506, USA;
| | - Tamal Saha
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka 1212, Bangladesh;
| |
Collapse
|
50
|
Chen R, Nie M, Jiang Y, Wu S, Wu J, Qiu D, Wu Y, Yuan Q, Wang S, Jiang Y, Zhang T. A respiratory mucosal vaccine based on chitosan/aluminum adjuvant induces both mucosal and systemic immune responses. Int J Pharm 2025; 670:125168. [PMID: 39756594 DOI: 10.1016/j.ijpharm.2025.125168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The respiratory mucosa serves as a critical barrier against the invasion of pathogens. Effective mucosal vaccines are essential for enhancing local immunity. However, there is an urgent need to develop new mucosal adjuvants. Chitosan is preferred as a mucosal adjuvant due to its mucosal adhesion and immunostimulatory properties. In this work, a novel mucosal adjuvant was synthesized by combining nano-aluminum hydroxide and chitosan (Al-CS), formulating a particle size approximately 1.5 μm. In vitro assays revealed that Al-CS notably promotes antigen uptake by enhancing activation and maturation of dendritic cells and macrophages. Furthermore, in vivo experiments indicated that Al-CS could extend antigen release duration, facilitate immune cell migration to the lungs, stimulates antigen-presenting cell maturation, enhances antigen presentation and significantly improves both humoral and cellular immunity as well as B/T cell memory differentiation. The immunological potential of Al-CS exceeds that of either aluminum or chitosan alone, making it a promising and safe adjuvant for the advancement of mucosal vaccine carrier systems.
Collapse
Affiliation(s)
- Ruitong Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yuetong Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuyu Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Junwei Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Dekui Qiu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangtao Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China
| | - Shaojuan Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Yao Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health (School of Life Science), Xiamen University, Xiamen, Fujian 351002, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|