101
|
Aizawa T. Socio-economic gradients in hypertension and diabetes management amid the COVID-19 pandemic in India. PLoS One 2025; 20:e0315867. [PMID: 40173110 PMCID: PMC11964223 DOI: 10.1371/journal.pone.0315867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/02/2024] [Indexed: 04/04/2025] Open
Abstract
This study examines socio-economic inequalities in the prevalence and treatment of hypertension and diabetes among adults in India, utilising data from the National Family Health Survey (NFHS) collected before and during the COVID-19 pandemic. Disparities associated with individual demographic and socio-economic characteristics are measured, with the level of inequality quantified using the dissimilarity index and contributing factors analysed through decomposition analysis. The results reveal significant socio-economic gradients, with wealthier individuals more likely to have elevated blood pressure and blood glucose levels and to treat them. Socio-economic gradients in treatment are even steeper among middle-aged groups during the pandemic. These wealth- and education-related disparities become more pronounced with age. This study highlights the need for targeted interventions and policies to address socio-economic disparities in access to essential care for socio-economically disadvantaged populations.
Collapse
Affiliation(s)
- Toshiaki Aizawa
- Graduate School of Economics and Business, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
102
|
Berhanu N, Hirpa E, Mohammed H, Legesse A, Deresse G, Assefa E, Tesgera T, Akalu M, Abayneh T, Bayissa B, Tesfaw L, Birhanu K, Gelaye E. Isolation, molecular identification, and phylogenetic analysis of infectious bronchitis virus from commercial chicken farms in Mekele and Bishoftu, Ethiopia, 2023-2024. Virol J 2025; 22:90. [PMID: 40176128 PMCID: PMC11963663 DOI: 10.1186/s12985-025-02639-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/22/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Avian infectious bronchitis (IB) is a highly contagious respiratory disease that affects the poultry industry globally. The disease is caused by avian infectious bronchitis virus (IBV), member of the genus Gammacoronavirus. In Ethiopia, IBV has been reported in both commercial and backyard chickens based on clinical observation. The objectives of this study were to isolate the virus, conduct molecular based identification, and phylogenetic analysis of the circulating IBV isolates. METHODS AND MATERIALS A cross-sectional study was conducted between November 2023 and May 2024 in Mekele and Bishoftu, Ethiopia. A total of 49 clinical samples were collected, comprising 12 tissue samples and 39 pooled swab samples. Of these, 6 samples-specifically, 5 swab samples and 1 tissue sample-tested positive for infectious bronchitis virus (IBV) through virus-specific conventional RT-PCR and real-time PCR. Nested PCR was performed using serotype-specific primers. The purified PCR products, which targeted the spike glycoprotein S1 subunit gene and the 3' UTR of the IBV, were sequenced, followed by phylogenetic tree analysis. RESULTS The six positive samples propagated into specific pathogen free embryonated eggs and exhibited characteristic IBV lesions and mortality observed over five consecutive passages. IBV isolates from Bishoftu (n = 4) and Mekele (n = 2) were amplified using one-step RT-PCR to target 466 bp of the S1 subunit gene and 433 bp of the 3'UTR. A BLAST search on the S1 partial gene and 3'UTR sequences, nested PCR, and phylogenetic analysis revealed that the present IBV isolates are genetically similar to the Massachusetts serotype. The S1 gene sequences of the five IBV isolates were deposited in GenBank with accession numbers PQ389500 to PQ389504. CONCLUSIONS This is the first detailed study on IB virus isolation, molecular detection, sequencing, and phylogenetic analysis in Ethiopia. The findings revealed that the outbreaks were caused by the IB virus, which created a serious health risk and economic losses in the chicken industry. To the author's knowledge, this is the first comprehensive study on the isolation and genetic analysis of IBV in Ethiopia. Further research on the economic impact of IBV in chicken production, farm biosecurity, serotyping of circulating IB virus, and vaccine development based on the local serotypes is recommended.
Collapse
Affiliation(s)
- Nigusu Berhanu
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia.
| | - Eyob Hirpa
- College of Veterinary Medicine and Agriculture, Department of Microbiology, Immunology and Veterinary Public Health, Addis Ababa University, P.O. Box 34, Bishoftu, Ethiopia
| | - Hawa Mohammed
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Abinet Legesse
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Getaw Deresse
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Eyob Assefa
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Takele Tesgera
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Mirtneh Akalu
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Takele Abayneh
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Berecha Bayissa
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Liyuwork Tesfaw
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Kenaw Birhanu
- Research and Development Directorate, National Veterinary Institute (NVI), P.O. Box 19, Bishoftu, Ethiopia
| | - Esayas Gelaye
- Emergency Centre for Transboundary Animal Diseases (ECTAD), Food and Agriculture Organization of the United Nations (FAO), P.O. Box 5536, Addis Ababa, Ethiopia.
| |
Collapse
|
103
|
Fatih MT, Mahmood MA, Mahmood MK, Tahir AI, Kurda HA, Abdulghafor MA, Qadir BH, Noori ZF. Post-COVID-19 Exacerbation of a Stable Fibrous Dysplasia: A Case Report. Clin Case Rep 2025; 13:e70474. [PMID: 40270775 PMCID: PMC12014514 DOI: 10.1002/ccr3.70474] [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: 11/12/2024] [Revised: 02/03/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
Fibrous dysplasia (FD) is a rare, benign fibro-osseous lesion characterized by replacement of normal bone with extensive fibrous stroma due to abnormalities in osteoblast differentiation. After puberty and during adulthood, FD lesions usually become quiescent. However, some cases show signs of regrowth and reactivation. Here, we report a previously stable maxillary FD case in a 32-year-old man reactivated after a mild COVID-19 infection. We hypothesize that SARS-CoV-2 may utilize diverse mechanisms to induce tumor/cancer in multiple organs, including initiating inflammatory cascades and modifying tumor-suppressing pathways. The capacity of SARS-CoV-2 to enhance the expression of proinflammatory and tumorigenic molecules necessitates further research to ascertain any correlation between this viral infection and FD or other similar diseases.
Collapse
Affiliation(s)
- Mohammed Taib Fatih
- Department of DentistryKomar University of Science and TechnologySulaimaniIraq
| | | | | | | | | | | | - Balen Hamid Qadir
- Department of DentistryKomar University of Science and TechnologySulaimaniIraq
| | - Zana Fuad Noori
- Dentistry CollegeAmerican University of Iraq‐Sulaimani AUISSulaimaniIraq
| |
Collapse
|
104
|
Sandhya K, Palanisamy PR, Sandhya R. COVID-19 pandemic versus six "A"s of health care services - accessibility, affordability, availability, appropriateness, adequacy, and adherence. J Family Med Prim Care 2025; 14:1320-1324. [PMID: 40396084 PMCID: PMC12088546 DOI: 10.4103/jfmpc.jfmpc_1535_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/18/2024] [Accepted: 10/26/2024] [Indexed: 05/22/2025] Open
Abstract
Background and Rationale COVID-19 pandemic resulted in a huge setback in health care sector causing morbidity, mortality, lack of manpower, resources, in hospitals. Apart from the direct effects of the disease, the indirect effects include, lockdowns, restriction of movement of people for food, work, medicines, etc. There are certain non-communicable diseases for which lifelong treatment and continuation of care is required and the patients suffering from these diseases were also highly affected during pandemic. The major pillars of health care services are accessibility, affordability, availability appropriateness, adequacy and adherence and it is true that the pandemic had its impact on all these pillars. Methodology A prospective cross sectional study in which 284 patients with NCD answered a questionaire and the responses were analysed. Results There was a lack of accessibility (48%), affordability (42%) availability (54%) appropriateness and 71% of population took self-medication during the period. There was discontinuity in 16% of the population and the reasons for discontinuation include depression, lack of motivation, forgetfulness, financial crisis etc. Conclusion The pandemic exerted a greater influence on saturation of health care services by increasing the mortality, morbidity and as a consequence decreased the health care services to communicable/non-communicable diseases. Our study shows the need and demand for robust health care services during critical times.
Collapse
Affiliation(s)
- Kalidas Sandhya
- Department of Pharmacology, JIPMER, Karaikal, Pondicherry, India
| | | | - R. Sandhya
- Department of Pharmacology, JIPMER, Karaikal, Pondicherry, India
| |
Collapse
|
105
|
Gondelaud F, Bignon C, Ptchelkine D, Carrière F, Longhi S. A conserved motif in Henipavirus P/V/W proteins drives the fibrillation of the W protein from Hendra virus. Protein Sci 2025; 34:e70085. [PMID: 40100133 PMCID: PMC11917119 DOI: 10.1002/pro.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 03/20/2025]
Abstract
The Hendra (HeV) and Nipah (NiV) viruses are high-priority, biosafety level-4 pathogens that cause fatal neurological and respiratory disease. Their P gene encodes not only the P protein, an essential polymerase cofactor, but also the virulence factors V and W. We previously showed that the W protein of HeV (WHeV) forms amyloid-like fibrils and that one of its subdomains, PNT3, fibrillates in isolation. However, the fibrillation kinetics is much faster in the case of the full-length WHeV compared to PNT3, suggesting that another WHeV region contributes to the fibrillation process. In this work, we identified the region spanning residues 2-110 (PNT1) as the crucial region implicated in WHeV fibrillation. Through site-directed mutagenesis, combined with thioflavin T binding experiments and negative-staining transmission electron microscopy, we showed that a predicted cryptic amyloidogenic region (CAR) within PNT1 is the main driver of fibrillation and deciphered the underlying molecular mechanism. Using FTIR, we showed that PNT1 fibrils are enriched in cross β-sheets. Sequence alignment revealed conservation of the CAR across the Henipavirus genus and enabled the identification of a hitherto never reported pro-amyloidogenic motif. The ability to form fibrils was experimentally shown to be a common property shared by Henipavirus PNT1 proteins. Overall, this study sheds light on the molecular mechanisms underlying WHeV fibrillation and calls for future studies aimed at exploring the relevance of the newly identified pro-amyloidogenic motif as a valuable target for antiviral approaches.
Collapse
Affiliation(s)
- Frank Gondelaud
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Christophe Bignon
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Denis Ptchelkine
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Frédéric Carrière
- Aix Marseille Univ, CNRS, UMR7281 Bioénergétique et Ingénierie des ProtéinesMarseilleFrance
| | - Sonia Longhi
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| |
Collapse
|
106
|
Chen Y, Dong Y, Wei S, Gao X, Li W, Zhao P. Genomic Integration of Hepatitis B Virus Into Human Hepatocytes in Early Childhood Cirrhosis. Liver Int 2025; 45:e70080. [PMID: 40130949 DOI: 10.1111/liv.70080] [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: 05/08/2024] [Revised: 02/09/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND AND AIMS Hepatitis B virus (HBV) remains a major global health problem. HBV DNA can be integrated into the human chromosomes. The integration in young cirrhotic chronic hepatitis B children has not been explored. This study aims to investigate HBV DNA integration in early childhood cirrhosis. METHODS Biopsy liver specimens from cirrhotic and matched non-cirrhotic chronic hepatitis B children were collected. HBV DNA integration was detected through targeted HBV DNA fragment capture sequencing. RESULTS Twenty cirrhotic and 20 non-cirrhotic children with chronic hepatitis B were included in the study. The cirrhotic group included 14 males and 6 females, and the non-cirrhotic group included 13 males and 7 females. Compared to non-cirrhotic children, cirrhotic children had lower serum HBsAg quantification (p = 0.001). The median number of HBV integrants in the cirrhotic group was 59 and that in the non-cirrhotic group was 98. No significant difference existed between the two groups (p = 0.529). In the multivariate linear regression analysis, serum HBV DNA level was correlated with the number of HBV integrants (p < 0.001, R2 = 0.322). Six differential intragenic high-frequency viral integration sites in cirrhotic children were revealed, all of which have protein-coding functions. CONCLUSION Several frequently integrated genes were observed in early childhood cirrhosis. Detailed associations between genetic alterations induced by HBV integration and early childhood cirrhosis need further exploration.
Collapse
Affiliation(s)
- Ying Chen
- Department of Clinical Laboratory, 962nd Hospital of PLA Joint Logistic Support Force, Harbin, Heilongjiang Province, China
| | - Yi Dong
- The Fifth Medical Center (formerly Beijing 302 Hospital), Chinese PLA General Hospital, Beijing, China
| | - Shizhang Wei
- Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Gao
- The Fifth Medical Center (formerly Beijing 302 Hospital), Chinese PLA General Hospital, Beijing, China
| | - Weijie Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Pan Zhao
- The Fifth Medical Center (formerly Beijing 302 Hospital), Chinese PLA General Hospital, Beijing, China
- Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
107
|
Thiruvaiyaru A, Mattila S, Sadeghi M, Naumenko K, Merits A, Varjosalo M, Ahola T. Proximity interactome of alphavirus replicase component nsP3 includes proviral host factors eIF4G and AHNAK. PLoS Pathog 2025; 21:e1013050. [PMID: 40193402 PMCID: PMC12005498 DOI: 10.1371/journal.ppat.1013050] [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: 11/14/2024] [Revised: 04/17/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025] Open
Abstract
All positive-strand RNA viruses replicate their genomes in association with modified intracellular membranes, inducing either membrane invaginations termed spherules, or double-membrane vesicles. Alphaviruses encode four non-structural proteins nsP1-nsP4, all of which are essential for RNA replication and spherule formation. To understand the host factors associated with the replication complex, we fused the efficient biotin ligase miniTurbo with Semliki Forest virus (SFV) nsP3, which is located on the cytoplasmic surface of the spherules. We characterized the proximal proteome of nsP3 in three cell lines, including cells unable to form stress granules, and identified >300 host proteins constituting the microenvironment of nsP3. These included all the nsPs, as well as several previously characterized nsP3 binding proteins. However, the majority of the identified interactors had no previously identified roles in alphavirus replication, including 39 of the top 50 interacting proteins. The most prominent biological processes involving the proximal proteins were nucleic acid metabolism, translational regulation, cytoskeletal rearrangement and membrane remodeling. siRNA silencing confirmed six novel proviral factors, USP10, AHNAK, eIF4G1, SH3GL1, XAB2 and ANKRD17, which are associated with distinct cellular functions. All of these except SH3GL1 were also important for the replication of chikungunya virus. We discovered that the small molecule 4E1RCat, which inhibits the interaction between the canonical translation initiation factors eIF4G and eIF4E, exhibits antiviral activity against SFV. Since the same molecule was previously found to inhibit coronaviruses, this suggest the possibility that translation initiation factors could be considered as targets for broadly acting antivirals.
Collapse
Affiliation(s)
- Aditya Thiruvaiyaru
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Sari Mattila
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Mohammadreza Sadeghi
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | | | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tero Ahola
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| |
Collapse
|
108
|
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
|
109
|
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] [Accepted: 11/05/2024] [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
|
110
|
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 PMCID: PMC12148012 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] [Grants] [Track Full Text] [Download PDF] [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
|
111
|
Wang X, He J, Sun M, Wang S, Qu J, Shi H, Rao B. High-dose vitamin C as a metabolic treatment of cancer: a new dimension in the era of adjuvant and intensive therapy. Clin Transl Oncol 2025; 27:1366-1382. [PMID: 39259387 DOI: 10.1007/s12094-024-03553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/04/2024] [Indexed: 09/13/2024]
Abstract
The anti-cancer mechanism of High-dose Vitamin C (HDVC) is mainly to participate in the Fenton reaction, hydroxylation reaction, and epigenetic modification, which leads to the energy crisis, metabolic collapse, and severe peroxidation stress that results in the proliferation inhibition or death of cancer cells. However, the mainstream view is that HDVC does not significantly improve cancer treatment outcomes. In clinical work and scientific research, we found that some drugs or therapies can significantly improve the anti-cancer effects of HDVC, such as PD-1 inhibitors that can increase the anti-cancer effects of cancerous HDVC by nearly three times. Here, the adjuvant and intensive therapy and synergistic mechanisms including HDVC combined application of chemoradiotherapies multi-vitamins, targeted drugs, immunotherapies, and oncolytic virus are discussed in detail. Adjuvant and intensive therapy of HDVC can significantly improve the therapeutic effect of HDVC in the metabolic treatment of cancer, but more clinical evidence is needed to support its clinical application.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jia He
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shiwan Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Jinxiu Qu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Hanping Shi
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| | - Benqiang Rao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Center of Metabolism and Nutrition of Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
- Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China.
| |
Collapse
|
112
|
Sharma V, Manchester L, Holstein M, Xu X, Ghose S, Heldt CL. Impact of Eco-Friendly Surfactant Structure and Class on Enveloped Virus Inactivation. Biotechnol J 2025; 20:e70023. [PMID: 40285390 DOI: 10.1002/biot.70023] [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: 11/27/2024] [Revised: 03/12/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Sustainable and effective strategies for virus inactivation are crucial for ensuring the safety and quality of biological products. The European Union's (EU) 2021 ban on Triton X-100 for viral inactivation in biomanufacturing has pushed the field to find sustainable alternatives with equal effectiveness. We aim to increase the sustainability of biopharmaceutical production by ensuring the effectiveness of eco-friendly surfactant-mediated virus inactivation by comparing the antiviral efficacy of Triton X-100 to glucosides and amine oxides. RESULTS Surfactants were evaluated for antiviral efficacy against herpes viruses, SuHV and HSV, and the retrovirus XMuLV. The surfactants demonstrated equivalent or superior inactivation efficacy compared to Triton X-100. Herpes viruses were inactivated similarly with all surfactants. For XMuLV, surfactants with longer alkyl chains achieved maximum log reduction values (LRV) at 1x CMC, outperforming Triton X-100, which required 2x CMC for comparable efficacy. Surfactants with bulky headgroups, such as LAPAO, showed lower efficacy against XMuLV. At a salt concentration of 2 M ionic strength, the antiviral efficacy of Triton X-100 and TDAO decreased for the herpes viruses. Variability in inactivation was observed among the surfactants at 0.5x CMC, indicating that surfactant characteristics influence their antiviral performance below CMC. CONCLUSIONS Adding salt enhanced the antiviral efficacy of surfactants by lowering their CMC while maintaining consistent virus inactivation. Among the surfactants tested, the glucoside with a longer tail, n-nonyl-β-D-glucoside (NG), emerged as the most robust and could function as an eco-friendly surfactant for virus inactivation in bioprocessing. For NG, virus inactivation was independent of all variables tested.
Collapse
Affiliation(s)
- Vaishali Sharma
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, USA
- Health Research Institute, Michigan Technological University, Houghton, Michigan, USA
| | - Lynn Manchester
- Department of Chemical Engineering, Michigan Technological University, Houghton, Michigan, USA
| | - Melissa Holstein
- Biologics Process Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Xuankuo Xu
- Biologics Process Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Sanchayita Ghose
- Biologics Process Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Caryn L Heldt
- Health Research Institute, Michigan Technological University, Houghton, Michigan, USA
- Department of Chemical Engineering, Michigan Technological University, Houghton, Michigan, USA
| |
Collapse
|
113
|
Ravi V, Khare K, Mohite R, Mishra P, Halder S, Shukla R, Liu CSC, Yadav A, Soni J, Kanika, Chaudhary K, Neha, Tarai B, Budhiraja S, Khosla P, Sethi T, Imran M, Pandey R. Genomic hotspots in the DENV-2 serotype (E, NS4B, and NS5 genes) are associated with dengue disease severity in the endemic region of India. PLoS Negl Trop Dis 2025; 19:e0013034. [PMID: 40299925 PMCID: PMC12040166 DOI: 10.1371/journal.pntd.0013034] [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: 12/30/2024] [Accepted: 04/04/2025] [Indexed: 05/01/2025] Open
Abstract
Dengue virus (DENV) infection manifests a wide spectrum of clinical outcomes, ranging from mild fever to severe and potentially fatal disease, yet the factors driving this variability remain poorly understood. This study aims to unravel the relationship between clinical manifestations of dengue and the genetic diversity of the virus, providing insights into the genomic variability driving disease severity. To achieve this, serum samples were collected during a dengue outbreak in National Capital Region-Delhi, India, from June to November 2023. Serotyping of RNA isolated from 4,045 clinical serum samples revealed DENV-2 as the predominant serotype in circulation (n = 3702). Whole-genome sequencing for 3702 clinical samples was performed using Oxford Nanopore Technology (ONT) further yielding 3254 DENV-2 genomes with >50% coverage. However, all of them identified the cosmopolitan genotype of DENV-2, forming a distinct monophyletic cluster in the global phylogenetic tree. Comprehensive variant analysis uncovered 1,618,158 single nucleotide variations (SNVs) across the sequenced DENV-2 population. The clinico-genomic approach carried out in 1294 samples, mild (n = 473), moderate (n = 405) and clinically severe (n = 416), reveals a significant burden of SNVs in various genomic regions linked to differential disease outcomes. Statistical analyses, including Fisher's exact test and phi-correlation, identified hotspot regions in the Envelope (E), NS4B, and NS5 genes, where SNVs were strongly associated with mild and clinically severe phenotypes, providing insights into the genomic determinants of disease severity. Interestingly, the clustering of severity-associated SNVs in these genomic hotspot regions highlights their potential as therapeutic targets within the DENV genome. These findings offer a promising direction for developing early mitigation strategies and targeted interventions to manage the progression of severe DENV infections.
Collapse
Affiliation(s)
- Varsha Ravi
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
- Indraprastha Institute of Information Technology, Delhi, India
| | - Kriti Khare
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ramakant Mohite
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Pallavi Mishra
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Sayanti Halder
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Richa Shukla
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Chinky Shiu Chen Liu
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Aanchal Yadav
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jyoti Soni
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kanika
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Komal Chaudhary
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Max Super Speciality Hospital (A Unit of Devki Devi Foundation), Max Healthcare, Delhi, India,
| | - Neha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Max Super Speciality Hospital (A Unit of Devki Devi Foundation), Max Healthcare, Delhi, India,
| | - Bansidhar Tarai
- Max Super Speciality Hospital (A Unit of Devki Devi Foundation), Max Healthcare, Delhi, India,
| | - Sandeep Budhiraja
- Max Super Speciality Hospital (A Unit of Devki Devi Foundation), Max Healthcare, Delhi, India,
| | | | | | - Md Imran
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
| | - Rajesh Pandey
- Division of Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
114
|
Mitchell CB, Simmons J, Hodo CL, Neal SJ, Chitta S, Vo C, Bakshi K, Juliani J, Fenske J, Cleveland DC, Cleveland JD. Early Results of an Infant Model of Orthotopic Cardiac Xenotransplantation. J Heart Lung Transplant 2025; 44:503-510. [PMID: 39778609 PMCID: PMC12025259 DOI: 10.1016/j.healun.2024.12.011] [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: 08/15/2024] [Revised: 11/26/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Genetically engineered porcine hearts may have an application for infants in need of a bridge to cardiac allotransplantation. The current animal model that resulted in 2 human applications has been validated in adult non-human primates only. We sought to create an infant animal model of life sustaining cardiac xenotransplantation to understand limitations specific to this age group. METHODS We performed 11 orthotopic cardiac xenotransplants from genetically modified infantile pigs into size-matched baboons (Papio spp). Porcine grafts were preserved using a modified Del Nido solution. Protocolized post-operative care and outcomes were tracked with invasive monitoring, echocardiogram, and serial chemistries (including a 7-cytokine panel). RESULTS Mean ischemic time was 52.1 +/- 13.9 min. All porcine hearts separated from bypass in normal sinus rhythm with normal systolic function documented by echocardiogram at chest closure and again at 24 h. In the first 48 post-operative hours, mean vasoactive inotropic score for the recipients was 9.6 +/- 3.5. Survival >3months was achieved in 6 animals. Five animals succumbed early (<7days) either due to errors in care (n=2) or pulmonary complications (n=3) confirmed on chest radiograph and necropsy. Cytokine levels objectively increased following xenograft implant but were not significantly different between survivors and non-survivors. CONCLUSIONS In a non-human primate model of infant orthotopic cardiac xenotransplantation, cardiac function does not hinder early peri-operative survival. Instead, pulmonary edema and pleural effusions in the setting of systemic inflammation preclude clinical progression. Targeted therapies are necessary to encourage prolonged survival.
Collapse
Affiliation(s)
- Chace B Mitchell
- Division of Cardiac Surgery, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA
| | - Joe Simmons
- Michale E. Keeling Center for Comparative Medicine, MD Anderson Cancer Center, Bastrop, TX
| | - Carolyn L Hodo
- Michale E. Keeling Center for Comparative Medicine, MD Anderson Cancer Center, Bastrop, TX
| | - Sarah J Neal
- Michale E. Keeling Center for Comparative Medicine, MD Anderson Cancer Center, Bastrop, TX
| | - Sriram Chitta
- Michale E. Keeling Center for Comparative Medicine, MD Anderson Cancer Center, Bastrop, TX
| | - Clementine Vo
- Division of Pediatric Cardiac Anesthesiology, Department of Anesthesia Critical Care Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Kanwarpal Bakshi
- Division of Pediatric Cardiac Anesthesiology, Department of Anesthesia Critical Care Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Julie Juliani
- Division of Cardiac Perfusion, Department of Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA
| | - Julie Fenske
- Division of Cardiac Perfusion, Department of Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA
| | - David C Cleveland
- Division of Cardiac Surgery, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA
| | - John D Cleveland
- Division of Cardiac Surgery, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA.
| |
Collapse
|
115
|
Liu H, Ramirez BM, Wong TS, Weiss CM, Lloyd KCK, Gong Q, Coffey LL. Severe Acute Respiratory Syndrome Coronavirus 2 Variant Infection Dynamics and Pathogenesis in Transgenic K18-h ACE2 and Inbred Immunocompetent C57BL/6J Mice. Viruses 2025; 17:500. [PMID: 40284943 PMCID: PMC12031173 DOI: 10.3390/v17040500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
The global impact of the COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), persists in part due to the emergence of new variants. Understanding variant-specific infection dynamics and pathogenesis in murine models is crucial for identifying phenotypic changes and guiding the development of countermeasures. To address the limitations of earlier studies that investigated only a few variants or used small sample sizes, we evaluated clinical disease, infection kinetics, viral titers, cellular localization, and histopathologic changes in the lungs and brains of transgenic B6.Cg-Tg(K18-ACE2)2Prlmn/J ("K18") and corresponding genetic control (C57BL/6J) mice expressing human angiotensin-converting enzyme 2 (hACE2). Six SARS-CoV-2 variants were assessed: B.1 (WA1-like), alpha, beta, delta, omicron, and omicron XBB.1.5, using cohorts of ≥18 mice. Following intranasal inoculation with B.1, alpha, beta, or delta variants, K18 mice experienced rapid weight loss and reached euthanasia criteria by 5-6 days post-inoculation (dpi). In contrast, K18 mice inoculated with both omicron variants recovered to their starting weight within 4-6 dpi. Infectious SARS-CoV-2 was detected in the oropharynx at 1 and2 dpi, in the lungs at 2, 4, and 6 dpi, and in the brain at 4 and 6 dpi for all variants except omicron. SARS-CoV-2 nucleoprotein was detected, and interstitial pneumonia of varying severity was observed in K18 mice infected with all variants. Brain lesions were identified in mice infected with the B.1, beta, and delta variants 6 dpi. As K18 mice express hACE2 in the brain-a feature not present in humans-we also compared infection dynamics of three variants to those of a mouse-adapted WA1 strain in C57BL/6J mice lacking the human ACE2 gene. C57BL/6J mice did not experience lethal disease, exhibited milder pneumonia, and had no evidence of neuroinvasion despite similar infection kinetics to K18 mice. These findings demonstrate contrasting phenotypes across the two models and reduced tropism and pathology of omicron compared to earlier variants in both models. This comprehensive analysis of SARS-CoV-2 variants in two mouse models provides valuable insights for model and variant selection for future studies.
Collapse
Affiliation(s)
- Hongwei Liu
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Brianna M. Ramirez
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA 95616, USA; (B.M.R.); (Q.G.)
| | - Talia S. Wong
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Christopher M. Weiss
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Kevin C. K. Lloyd
- Mouse Biology Program, University of California, Davis, CA 95616, USA
- Department of Surgery, School of Medicine, University of California, Davis, CA 95616, USA
| | - Qizhi Gong
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA 95616, USA; (B.M.R.); (Q.G.)
| | - Lark L. Coffey
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
116
|
Morar R, Varga NI, Horhat DI, Mot IC, Balica NC, Tischer AA, Susan M, Susan R, Lighezan DL, Negrean RA. Primary Palatine Tonsil Non-Hodgkin Lymphoma in Western Romania: A Comparison of Lower-Stage and Advanced-Stage Disease. Hematol Rep 2025; 17:17. [PMID: 40277841 PMCID: PMC12026754 DOI: 10.3390/hematolrep17020017] [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: 02/08/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Limited data exist on primary palatine tonsil Non-Hodgkin lymphoma (NHL) from regions with constrained healthcare access. This study investigated this malignancy in Western and South-Western Romania, comparing lower-stage (Ann-Arbor I-III) and advanced-stage (IV) disease. Methods: A retrospective cohort study (2010-2019) at a tertiary referral hospital included 59 patients with primary palatine tonsil NHL. Data on demographics, clinical presentation, comorbidities (including viral hepatitis B/C), histology, International Prognostic Index (IPI) score, treatment, and outcomes were collected. Statistical comparisons between lower-stage (n = 26) and advanced-stage (n = 33) groups were performed. Results: A high proportion presented with advanced-stage disease (55.9%). The advanced-stage group had significantly more B symptoms (90.9% vs. 69.2%, p = 0.038) and elevated LDH levels (93.9% vs. 57.7%, p = 0.013). Viral hepatitis B and/or C infection was more frequent in advanced-stage disease (30.3% vs. 15.4%, p = 0.44). Combined chemoradiotherapy was more commonly used in lower-stage disease (38.46% vs. 12.12%, p = 0.019). There was no statistically significant difference in relapse rates between the groups. Conclusions: This study highlights the substantial burden of advanced-stage primary palatine tonsil NHL in Western Romania, suggesting a need for improved early detection. The association between viral hepatitis and advanced-stage, although not statistically significant, warrants further investigation. These findings may inform tailored management approaches in resource-constrained settings.
Collapse
Affiliation(s)
- Raluca Morar
- Doctoral School, Department of General Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (R.M.); (N.-I.V.)
| | - Norberth-Istvan Varga
- Doctoral School, Department of General Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (R.M.); (N.-I.V.)
| | - Delia Ioana Horhat
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (D.I.H.); (N.C.B.); (A.-A.T.)
| | - Ion Cristian Mot
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (D.I.H.); (N.C.B.); (A.-A.T.)
| | - Nicolae Constantin Balica
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (D.I.H.); (N.C.B.); (A.-A.T.)
| | - Alina-Andree Tischer
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (D.I.H.); (N.C.B.); (A.-A.T.)
| | - Monica Susan
- Department of Internal Medicine I, Centre for Preventive Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Razvan Susan
- Department of Family Medicine, Centre for Preventive Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Diana Luisa Lighezan
- Multidisciplinary Research Center for Malignant Hematological Diseases, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Rodica Anamaria Negrean
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| |
Collapse
|
117
|
Tanneti NS, Stillwell HA, Weiss SR. Human coronaviruses: activation and antagonism of innate immune responses. Microbiol Mol Biol Rev 2025; 89:e0001623. [PMID: 39699237 PMCID: PMC11948496 DOI: 10.1128/mmbr.00016-23] [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: 12/20/2024] Open
Abstract
SUMMARYHuman coronaviruses cause a range of respiratory diseases, from the common cold (HCoV-229E, HCoV-NL63, HCoV-OC43, and SARS-CoV-2) to lethal pneumonia (SARS-CoV, SARS-CoV-2, and MERS-CoV). Coronavirus interactions with host innate immune antiviral responses are an important determinant of disease outcome. This review compares the host's innate response to different human coronaviruses. Host antiviral defenses discussed in this review include frontline defenses against respiratory viruses in the nasal epithelium, early sensing of viral infection by innate immune effectors, double-stranded RNA and stress-induced antiviral pathways, and viral antagonism of innate immune responses conferred by conserved coronavirus nonstructural proteins and genus-specific accessory proteins. The common cold coronaviruses HCoV-229E and -NL63 induce robust interferon signaling and related innate immune pathways, SARS-CoV and SARS-CoV-2 induce intermediate levels of activation, and MERS-CoV shuts down these pathways almost completely.
Collapse
Affiliation(s)
- Nikhila S. Tanneti
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen A. Stillwell
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
118
|
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. [PMID: 40134845 PMCID: PMC11612881 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
|
119
|
Shamim L, Musharaf I, Nashwan AJ. Dexamethasone in coronavirus disease 2019 care: Dosage and utilization insights. World J Virol 2025; 14:98765. [PMID: 40134846 PMCID: PMC11612875 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
|
120
|
Varama A. Revisiting dexamethasone dosage in COVID-19 management. World J Virol 2025; 14:98359. [PMID: 40134844 PMCID: PMC11612871 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: 06/25/2024] [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
|
121
|
Sarker MS. Rhabdomyolysis-related acute kidney injury in COVID-19: A critical concern. World J Virol 2025; 14:100160. [PMID: 40134840 PMCID: PMC11612878 DOI: 10.5501/wjv.v14.i1.100160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [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
|
122
|
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. [PMID: 40134836 PMCID: PMC11612879 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
|
123
|
Moré JS, Serbena DR, de Camargo LGG, Clemente PA, dos Santos FS, Bonini JS. How the COVID-19 pandemic affected routine child vaccination: an integrative review. EINSTEIN-SAO PAULO 2025; 23:eRW1119. [PMID: 40136151 PMCID: PMC11991735 DOI: 10.31744/einstein_journal/2025rw1119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/04/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Child immunization plays a critical role in preventing numerous diseases. However, the COVID-19) pandemic has profoundly disrupted healthcare systems globally, including routine child vaccination programs. OBJECTIVE To provide an overview of the reduction in vaccine coverage among infants and children during the pandemic and analyze the potential impacts of decreased child immunization during this period. METHODS A comprehensive search was conducted using the MeSH terms "Child," "Vaccination", and "COVID-19," along with their synonyms. Systematic reviews published between March 11, 2020, and June 1, 2023, in Portuguese or English were included. Databases searched included PubMed, BVS (Biblioteca Virtual em Saúde), Embase, and Scopus. Two blinded independent reviewers performed the selection process, with conflicts resolved by a third reviewer. The AMSTAR-II tool was used to assess the methodological quality of the included studies. RESULTS Of the 1,534 eligible articles, only 13 addressed the pandemic's impact on children's vaccination coverage. Most studies involved multiple countries and reported a significant decrease in children's vaccination coverage due to the COVID-19 pandemic. CONCLUSION The findings were heterogeneous but consistently highlighted the substantial impact of the COVID-19 pandemic on routine vaccination coverage in most countries. Further research is needed to explore the epidemiological consequences of disruptions to vaccination schedules, potentially guiding public policies and raising awareness about the importance of adhering to health protection programs.
Collapse
Affiliation(s)
- Julia Stoeterau Moré
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| | - Daniel Rodrigo Serbena
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| | - Luiz Gustavo Gusson de Camargo
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| | - Pedro Augusto Clemente
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| | - Fernando Sluchensci dos Santos
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| | - Juliana Sartori Bonini
- Laboratório de Neurociências e ComportamentoUniversidade Estadual do Centro-OesteGuarapuavaPRBrazil Laboratório de Neurociências e Comportamento, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil.
| |
Collapse
|
124
|
Boukaira S, Madihi S, Bouafi H, Rchiad Z, Belkadi B, Benani A. Hepatitis C in North Africa: A Comprehensive Review of Epidemiology, Genotypic Diversity, and Hepatocellular Carcinoma. Adv Virol 2025; 2025:9927410. [PMID: 40165825 PMCID: PMC11957868 DOI: 10.1155/av/9927410] [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: 09/05/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Hepatitis C virus (HCV) is implicated in carcinogenic infections like hepatocellular carcinoma (HCC). Given that no HCV vaccine is currently available, comprehensive epidemiological understanding is crucial for devising effective prevention strategies. In North Africa, existing data on HCV infection and HCV-associated HCC are frequently outdated or limited to specific populations. This systematic review aims to offer new insights into the epidemiology of HCV infection, HCV genotype distribution, and HCV-related HCC in this region. We collected data from electronic databases: PubMed, ScienceDirect, ResearchGate, Google Scholar, and public health reports between 1989 and 2023. We reported the studies carried out in each country in general populations and in groups exposed to HCV infection. Our results show that HCV prevalence varies from 0.5% to 4.61% among the general populations in North African countries. HCV genotype 1 remains the most dominant in the Greater Maghreb region, while genotype 4 is the most dominant in the Nile Valley region. HCC incidence varies between the five countries, and HCV is responsible for 60% of cases, with male dominance. Egypt had the highest number of deaths from HCV-associated HCC. Other factors such as HBV, diabetes, and alcohol use are also responsible for HCC in North Africa. Urban growth and socioeconomic changes have impacted HCV prevalence in the North African region, especially among rural populations, and introduced new risks, such as coinfections and Type 2 diabetes. Here, we propose some recommendations for HCV control and management by patient category in North Africa.
Collapse
Affiliation(s)
- Samia Boukaira
- Department of Molecular Biology, Institut Pasteur du Maroc, Casablanca, Morocco
- Department of Microbiology and Molecular Biology, Faculté des Sciences, Université Mohammed V de Rabat, Rabat, Morocco
| | - Salma Madihi
- Department of Molecular Biology, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hind Bouafi
- Department of Human Genomics and Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Zineb Rchiad
- Department of Biological Sciences, Faculty of Medical Sciences, Université Mohammed VI Polytechnique EMINES, Benguerir, Morocco
| | - Bouchra Belkadi
- Department of Microbiology and Molecular Biology, Faculté des Sciences, Université Mohammed V de Rabat, Rabat, Morocco
| | - Abdelouaheb Benani
- Department of Molecular Biology, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
125
|
Hendawy AS, Sabra ANA, George MY, Rashad E, El-Demerdash E, Botros SS. The antifibrotic effect of Vildagliptin and Diaminodiphenyl Sulfone in murine schistosomiasis mansoni. Sci Rep 2025; 15:10084. [PMID: 40128243 PMCID: PMC11933376 DOI: 10.1038/s41598-025-91955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Schistosomiasis drastically affects human health, where S. mansoni-induced hepatic fibrosis remains a serious problem with no available drug yet. The current study aimed to evaluate the hepatoprotective effects of Vildagliptin (Vilda), Diaminodiphenyl Sulfone (DDS), and their combination (Vilda/DDS) against S. mansoni-induced hepatic fibrosis and elucidate their underlying molecular mechanisms. S.mansoni-infected mice were administered praziquantel (PZQ) for two consecutive days, or Vilda, DDS, and Vilda/DDS for 14 consecutive days. Schistosomiasis-induced hepatic fibrosis was assessed parasitologically, biochemically, and pathologically. Results revealed that Vilda, DDS, and Vida/DDS treatments significantly reduced worm count, oogram stages, ova count, and ameliorated the granulomatous inflammatory reactions and hepatotoxicity indices. Moreover, they enhanced hepatic Nrf2/HO-1 pathway with significant increasing SOD and reducing MDA levels. Furthermore, they significantly downregulated the hepatic TLR4/NF-κB and NLRP3 inflammasome pathways leading to a significant reduction in TNF-α and caspase-1 levels which is important in the activation of IL-1β and caspase-3. Notably, significant downregulation in hepatic TGF-β1, α-SMA, and MMP-9 expressions were also recorded. In conclusion, Vilda/DDS showed antioxidant, anti-inflammatory and antifibrotic activities in comparison to either Vilda or DDS alone against S. mansoni-induced hepatic fibrosis. Therefore, Vilda/DDS is a promising approach for managing S. mansoni infection, liver fibrosis, and associated disease morbidity.
Collapse
Affiliation(s)
- Amira S Hendawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, P.O. Box 30, Imbaba, Giza, 12411, Egypt
| | - Abdel-Nasser A Sabra
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, P.O. Box 30, Imbaba, Giza, 12411, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, 11566, Egypt
| | - Eman Rashad
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, 11566, Egypt.
| | - Sanaa S Botros
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, P.O. Box 30, Imbaba, Giza, 12411, Egypt
| |
Collapse
|
126
|
de Assumpção LECC, Romeo BGP, Guerra JCDC, Camargo LFA, Nagaoka MA, Amgarten DE, Dorlass EG, Petroni RC, Cardoso ACA, Ruiz RDM, Bittante CD, Teich VD, Pinho JRR, Doi AM. Case report: Persistent COVID-19 in a patient with B cell lymphoma refractory to antiviral treatment due to resistance to Remdesivir. IDCases 2025; 40:e02199. [PMID: 40226209 PMCID: PMC11986235 DOI: 10.1016/j.idcr.2025.e02199] [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: 08/12/2024] [Revised: 02/18/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Background There is a significant concern of the pandemic impact of SARS-CoV-2 infection in immunocompromised patients. These patients can develop long COVID-19 due to impairment of cellular and humoral immunity. On the other hand, prolonged infection can lead to mutations in the SARS CoV-2 genome that can impact on the resistance to antiviral therapy. Remdesivir cases have been reported in patients receiving antiviral drug treatment. Case presentation A 46-year-old male with previous mantle cell lymphoma resolved by autologous bone marrow transplantation without other comorbidities had SARS-CoV-2 detected in February 2022 and received the recommended antiviral treatment with Remdesivir. COVID-19 evolved in four months with worsening of the symptoms, despite an initial rapid improvements and high RT-PCR Ct values. The patient was relieved from hospital care stable and well but still maintaining positive test results. Conclusions the patient presented prolonged COVID-19 with persistence of virus detected by RT-PCR for several months. The strain sequenced presented a mutation different from all reported previously. Although it was no possible to sequence the initial strain without these mutations, our data suggests that immunocompromised patient with prolonged COVID-19 may serve as reservoir for strains of SARS-CoV-2 with resistant components in his genome.
Collapse
Affiliation(s)
| | | | | | - Luis Fernando Aranha Camargo
- School of Medicine, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
- Hospital Israelita Albert Einstein, Brazil
| | - Marcelo Akira Nagaoka
- School of Medicine, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | - André Mario Doi
- School of Medicine, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
- Hospital Israelita Albert Einstein, Brazil
| |
Collapse
|
127
|
Qiao L, Yuan H. Prognostic value of C-reactive protein levels in pulmonary infections: A systematic review and meta-analysis. Medicine (Baltimore) 2025; 104:e41722. [PMID: 40128046 PMCID: PMC11936577 DOI: 10.1097/md.0000000000041722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/12/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND C-reactive rotein (CRP) has been extensively studied as a biomarker that can predict mortality in patients with acute lung disease and our study aimed to elucidate the prognostic value of CRP levels for mortality in patients with various airway diseases, accounting for these differences and potential confounding factors accounts. METHODS An extensive literature search was conducted in several databases including PubMed, Embase, Web of Science, Scopus, and ProQuest to ensure the inclusion of up-to-date evidence from studies published between January 2019 and December 2024. Both fixed-effects and random-effects models were used to calculate pooled mean hazard ratios (HR) and odds ratios (OR) for mortality. RESULTS For mortality, the fixed effects model revealed a HR of 1.0065 (95% CI: 1.0054-1.0075, P < .0001), indicating a slightly increased risk of death associated with higher CRP levels. However, the random effects model, considering study heterogeneity, suggested an HR of 1.0488 (95% CI: 0.9978-1.1024, P = .0608), with significant heterogeneity (Q = 135.31, P < .0001). The OR analysis under the random effects model showed a more substantial increase in mortality risk with an OR of 1.2033 (95% CI: 1.0635-1.3614, P = .0033). Regarding ICU admissions and ventilation needs, substantial heterogeneity was also observed. The analysis did not find a statistically significant association between elevated CRP levels and ICU admission (OR = 1.1108, 95% CI: 0.9604-1.2847, P = .1568) or the necessity for ventilation (OR = 1.8981, 95% CI: 0.9651-3.7331, P = .0633), although both indicated trends towards increased risk. CONCLUSION CRP levels show a potential yet inconsistent association with mortality risk in patients with pulmonary infections. While elevated CRP levels suggest an increased risk of mortality, the results should be interpreted cautiously due to potential overestimation of the effect and the presence of publication bias.
Collapse
Affiliation(s)
- Li Qiao
- Intensive Care Unit, Peking University International Hospital, Beijing, China
| | - Hongxun Yuan
- Intensive Care Unit, Peking University International Hospital, Beijing, China
| |
Collapse
|
128
|
Simioni C, Sanz JM, Gafà R, Cenacchi G, Occhionorelli S, Passaro A, Neri LM. Increase of VEGF and Fibronectin expression and ultrastructural alterations of intercellular junctions in a swab negative patient after SARS-COV-2 infection. Virol J 2025; 22:82. [PMID: 40114185 PMCID: PMC11927238 DOI: 10.1186/s12985-025-02701-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND SARS-CoV-2 infection has been responsible of COrona VIrus Disease (COVID-19) pandemia and can cause a variety of symptoms including gastrointestinal disorders, abdominal pain and liver injury. The host receptor for SARS-CoV-2, ACE2, is expressed in gut and SARS-CoV-2 infection could induce vascular damage and immune system dysregulation, creating an inflammatory and hypercoagulable state, as widely described at the lung level. CASE PRESENTATION This work presents the case of a middle-aged Caucasian man admitted to the Hospital Emergency Department from the University Hospital of Ferrara (Italy), complaining of pain in the upper and middle region of the abdomen. The patient tested negative to the nose-oropharyngeal swab for SARS-CoV-2 four weeks after recovering from viral infection. The patient required resection of a segment of ileum and an ulcer of the bowel wall was recognized and sampled. Previous published results had confirmed the presence of the SARS-CoV-2 nucleocapsid protein, an increased human leukocyte antigen (HLA-G) and an altered morphology of microvilli in the ulcerated ileum of the patient when compared to the non-ulcerated ileum. The present study sought to deepen the consequences of SARS-CoV-2 infection. To this end, we evaluated the expression and co-expression of Vascular Endothelial Growth Factor (VEGF) and Fibronectin by immunohistochemical techniques. VEGF immunohistochemical expression was higher in the ulcer than in the control ileum sample and the non-ulcerated ileum areas and co-expressed with the SPIKE protein. Fibronectin staining was lower in control sample than in non-ulcerated and ulcerated ileum. Electron microscopy analysis showed alterations of the integrity of the intestinal barrier in the ulcerated area when compared to the non-ulcerated ileum or to the control sample. CONCLUSIONS Although the patient was tested negative to nose-oropharyngeal swab for SARS-CoV-2, the SPIKE protein was detected in his terminal ileum, especially in the ulcerated areas. The presence of the viral protein was also associated with an increase of VEGF and Fibronectin. In addition to vascular changes, the SARS-CoV-2 infection altered the junctional apparatus among epithelial cells, making the tissue even more fragile and thus susceptible to the entry of pathogens and the development of further infections.
Collapse
Affiliation(s)
- Carolina Simioni
- Department of Life Sciences and Biotechnology, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA)-Electron Microscopy Center, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy
| | - Juana Maria Sanz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy
- Integrated Activity Department of Onco-Hematology, University Hospital of Ferrara Arcispedale, Sant'Anna - Via Aldo Moro 8, Ferrara, I-44124, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, 40126, Italy
| | - Savino Occhionorelli
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy
- Emergency Surgery Department, University Hospital of Ferrara Arcispedale Sant'Anna, Via Aldo Moro 8, Ferrara, I-44124, Italy
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy.
| | - Luca Maria Neri
- Laboratory for Technologies of Advanced Therapies (LTTA)-Electron Microscopy Center, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy.
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, Ferrara, I-44121, Italy.
| |
Collapse
|
129
|
Garneau WM, Wang K, Liang T, Xu Y, Gladstone DE, Avery RK, D'Alessio FR, Robinson ML, Sahetya SK, Garibaldi BT, Gebo KA, Dioverti MV. Effect of antecedent B-cell depletion therapy for patients hospitalized with COVID-19 within a single health system: a propensity score analysis. Sci Rep 2025; 15:9647. [PMID: 40113850 PMCID: PMC11926165 DOI: 10.1038/s41598-025-94024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
B-cell depletion therapy is employed in a variety of clinical contexts from auto-immune diseases to malignancy. Prior research on patients with prior B-cell depletion treatment has suggested a mortality risk in patients hospitalized with COVID-19 however previous case-control studies have differed in their methods of patient comparison. Patients previously treated with B-cell-depletion hospitalized with COVID-19 were compared to matched controls in the Johns Hopkins Health System between March 1, 2020 and November 30, 2021. The primary outcome was 30-day all-cause mortality. Secondary outcomes included time to severe illness or death and time to clinical improvement. To eliminate bias due to imbalanced covariates, each patient who had previously received B-cell depletion therapy was matched with patients who had not received therapy based on age, sex, race, WHO severity score, admission date, COVID-19 specific treatment, and vaccination status. Propensity scores were calculated from a multivariable logistic regression model and performed on the matched sets, using B-cell depletion as the outcome, where the propensity score was the probability of receiving B-cell depletion therapy. The propensity score included matched covariates as well as smoking status, medical comorbidities, and vaccination status. Cox proportional-hazards regression models were applied on the matched sets to perform time to death, time to severe illness or death, and time to clinical improvement analyses. 50 patients were identified who had received B-cell depletion therapy and were compared to 186 matched controls. Patients treated with B-cell depletion experienced 30-day mortality of 6.0% compared to 3.8% in controls, adjusted hazard ratio (aHR) 1.45 (95% CI 0.30 to 6.95). B-cell-depleted patients experienced a longer time to clinical improvement, aHR 0.65 (95% CI 0.45-0.94). In this cohort, patients treated with B-cell depletion experienced a higher mortality rate compared to matched controls however this was not statistically significant. This group also experienced a prolonged time to clinical improvement based on WHO severity score.
Collapse
Affiliation(s)
- William M Garneau
- Division of Hospital Medicine, Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie 2nd Floor, Suite 249, Room 256, Baltimore, MD, 21287, USA.
| | - Kunbo Wang
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Tao Liang
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
- Division of Biostatistics and Bioinformatics, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Douglas E Gladstone
- Division of Hematology/Medical Oncology, Northwell Health, New Hyde Park, NY, USA
| | - Robin K Avery
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew L Robinson
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarina K Sahetya
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly A Gebo
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M Veronica Dioverti
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
130
|
Koutsi M, Pouliou M, Chatzopoulos D, Champezou L, Zagkas K, Vasilogianni M, Kouroukli A, Agelopoulos M. An evolutionarily conserved constellation of functional cis-elements programs the virus-responsive fate of the human (epi)genome. Nucleic Acids Res 2025; 53:gkaf207. [PMID: 40131776 PMCID: PMC11934927 DOI: 10.1093/nar/gkaf207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Human health depends on perplexing defensive cellular responses against microbial pathogens like Viruses. Despite the major effort undertaken, the (epi)genomic mechanisms that human cells utilize to tailor defensive gene expression programs against microbial attacks have remained inadequately understood, mainly due to a significant lack of recording of the in vivo functional cis-regulatory modules (CRMs) of the human genome. Here, we introduce the virus-responsive fate of the human (epi)genome as characterized in naïve and infected cells by functional genomics, computational biology, DNA evolution, and DNA Grammar and Syntax investigations. We discovered that multitudes of novel functional virus-responsive CRMs (vrCRMs) compose typical enhancers (tEs), super-enhancers (SEs), repetitive-DNA enhancers (rDEs), and stand-alone functional genomic stretches that grant human cells regulatory underpinnings for layering basal immunity and eliminating illogical/harmful defensive responses under homeostasis, yet stimulating virus-responsive genes and transposable elements (TEs) upon infection. Moreover, extensive epigenomic reprogramming of previously unknown SE landscapes marks the transition from naïve to antiviral human cell states and involves the functions of the antimicrobial transcription factors (TFs), including interferon response factor 3 (IRF3) and nuclear factor-κB (NF-κB), as well as coactivators and transcriptional apparatus, along with intensive modifications/alterations in histone marks and chromatin accessibility. Considering the polyphyletic evolutionary fingerprints of the composite DNA sequences of the vrCRMs assessed by TFs-STARR-seq, ranging from the animal to microbial kingdoms, the conserved features of antimicrobial TFs and chromatin complexes, and their pluripotent stimulus-induced activation, these findings shed light on how mammalian (epi)genomes evolved their functions to interpret the exogenous stress inflicted and program defensive transcriptional responses against microbial agents. Crucially, many known human short variants, e.g. single-nucleotide polymorphisms (SNPs), insertions, deletions etc., and quantitative trait loci (QTLs) linked to autoimmune diseases, such as multiple sclerosis (MS), systemic lupus erythematosus (SLE), Crohn's disease (CD) etc., were mapped within or vastly proximal (±2.5 kb) to the novel in vivo functional SEs and vrCRMs discovered, thus underscoring the impact of their (mal)functions on human physiology and disease development. Hence, we delved into the virus-responsive fate of the human (epi)genome and illuminated its architecture, function, evolutionary origins, and its significance for cellular homeostasis. These results allow us to chart the "Human hyper-Atlas of virus-infection", an integrated "molecular in silico" encyclopedia situated in the UCSC Genome Browser that benefits our mechanistic understanding of human infectious/(auto)immune diseases development and can facilitate the generation of in vivo preclinical animal models, drug design, and evolution of therapeutic applications.
Collapse
Affiliation(s)
- Marianna A Koutsi
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Marialena Pouliou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Dimitris Chatzopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Lydia Champezou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Konstantinos Zagkas
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Marili Vasilogianni
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Alexandra G Kouroukli
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Marios Agelopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| |
Collapse
|
131
|
Yan P, Yu X, Chen Z, Lan L, Kang J, Zhao B, Liu D. Assessing the consistency of FIB-4, APRI, and GPR in evaluating significant liver fibrosis and cirrhosis in COVID-19 patients with concurrent liver diseases. BMC Gastroenterol 2025; 25:191. [PMID: 40114058 PMCID: PMC11927168 DOI: 10.1186/s12876-025-03770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE This study investigated the consistency of the FIB-4, APRI, and GPR indices in assessing significant liver fibrosis and cirrhosis in patients with Coronavirus Disease 2019(COVID-19) who also suffer from various liver diseases, providing references for the clinical selection and application for non-invasive assessment methods. METHODS The study evaluated 744 COVID-19 patients with coexisting liver diseases: 508 cases with non-alcoholic fatty liver disease (NAFLD), 158 cases with chronic hepatitis B (CHB), and 78 cases with a combination of both ailments. FIB-4, APRI, and GPR were employed to assess significant liver fibrosis and cirrhosis. Concordance among the methods was determined using Kappa analysis, and receiver operating characteristic (ROC) curves helped identify the optimal cutoff values for each index. RESULTS For COVID-19 patients with NAFLD, Kappa values for significant liver fibrosis were 0.81, 0.90, 0.80, and 0.79, and for cirrhosis, they were 0.88, 0.97,0.88, and 0.88, respectively (all p < 0.05). Among those with CHB, Kappa values were 0.81, 0.81, 0.83, and 0.75 for fibrosis, and0.87, 0.91, 0.88, and 0.92 for cirrhosis (all p < 0.05). In patients with coexisting liver diseases, the values were 0.87, 0.86, 0.86, and 0.78 for fibrosis, and 0.67, 0.69, 0.54, and 0.81for cirrhosis (all p < 0.05). Linear trend analysis revealed significant relationships between FIB-4 values, APRI values, GPR values, and the severity of COVID-19 (χ2 trend: 15.205,35.114, and 13.973, respectively, all p < 0.001), between FIB-4 values and APRI values and the coronavirus negative conversion time (all p < 0.05) in COVID-19 with NAFLD, and between FIB-4 values and GPR values and the coronavirus negative conversion time in patients with COVID-19 with CHB(all p < 0.05). CONCLUSION Using the current cutoff values, the non-invasive assessments demonstrated almost perfect consistency in evaluating significant liver fibrosis and cirrhosis in COVID-19 patients with liver diseases, though FIB-4 and GPR showed moderate consistency in cirrhosis evaluation in patients with coexisting liver conditions. Moreover, it also indicated that increased liver fibrosis correlates with more severe COVID-19 and prolonged coronavirus negative conversion time.
Collapse
Affiliation(s)
- Pan Yan
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan Province, 610500, China
| | - Xiaoping Yu
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan Province, 610106, China
| | - Zhu Chen
- Department of Drug Clinical Trial Center, Public Health Clinical Centre of Chengdu, Chengdu, Sichuan Province, 610060, China
| | - Lijuan Lan
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, Sichuan Province, 610060, China
| | - Jun Kang
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, Sichuan Province, 610060, China
| | - Bennan Zhao
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, Sichuan Province, 610060, China
| | - Dafeng Liu
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, Sichuan Province, 610060, China.
- , No.377 Jingming Road, Jinjiang District, Chengdu City, Sichuan Province Chengdu, 610060, China.
| |
Collapse
|
132
|
Lan Y, Xie Z, Cao Q, Mai C, Cheng L, Ning H, Huang Q, Li Z, Cui P, Ye L, Liang H, Huang J. Severe Anemia Is an Independent Risk Factor for In-Hospital Death in Persons Living with HIV in Southern China: A Retrospective Cohort Study. AIDS Res Hum Retroviruses 2025. [PMID: 40104875 DOI: 10.1089/aid.2024.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
To assess the impact of anemia severity during antiretroviral therapy (ART) on in-hospital mortality among persons living with HIV. We conducted a retrospective cohort study of hospitalized persons living with HIV at the Fourth People's Hospital of Nanning, Guangxi, China, from 2018 to 2020. Kaplan-Meier analysis was used to calculate cumulative mortality rates. The Cox proportional hazards model, 1:1:1 propensity score matching (PSM), and three-group inverse probability of treatment weighting (IPTW) were used to assess the impact of anemia severity on mortality in hospitalized persons living with HIV. A total of 2,217 hospitalized persons living with HIV were included, among whom 409 (18.4%) had anemia: 50 (2.3%) with mild anemia, 174 (7.8%) with moderate anemia, and 185 (8.3%) with severe anemia. Among all AIDS-related complications, patients with severe anemia had a higher mortality rate [20.34/100 person-months, 95% confidence interval (CI): 13.29-27.39], significantly higher than that of persons living with HIV without anemia (7.74/100 person-months, 95% CI: 6.02-9.45); the adjusted hazard ratio (AHR) was 2.422, with a 95% CI of (1.500, 3.913). After PSM and IPTW analyses, results were similar, with PSM (AHR: 4.745, 95% CI: 2.231-10.091) and IPTW (AHR: 1.920, 95% CI: 1.146-3.216). Patients with CD4+ T cell counts below 350 per μL and severe anemia had an increased mortality risk. Severe anemia is an independent risk factor for in-hospital death in persons living with HIV in Southern China. The importance of timely identification and assessment of anemia severity during ART and prompt treatment to correct anemia, which is crucial for improving anemia burden and prognosis for persons living with HIV.
Collapse
Affiliation(s)
- Yingji Lan
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhiman Xie
- Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Qin Cao
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Chen Mai
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Liping Cheng
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Huan Ning
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Qianqian Huang
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhuoxin Li
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| | - Ping Cui
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Li Ye
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Hao Liang
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Jiegang Huang
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
133
|
Esskhayry S, Benamri I, Lamzouri A, Kaissi O, Fissoune R, Moussa A, Radouani F. Adoption of an in-silico analysis approach to assess the functional and structural impacts of rpoB-encoded protein mutations on Chlamydia pneumoniae sensitivity to antibiotics. BMC Microbiol 2025; 25:157. [PMID: 40102727 PMCID: PMC11921668 DOI: 10.1186/s12866-025-03860-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Antibiotics are frequently used to treat infections caused by Chlamydia pneumoniae; an obligate intracellular gram-negative bacterium commonly associated with respiratory diseases. However, improper or overuse of these drugs has raised concerns about the development of antibiotic resistance, which poses a significant global health challenge. Previous studies have revealed a link between mutations in the rpoB-encoded protein of C. pneumoniae and antibiotic resistance. This study assessed these mutations via various bioinformatics tools to predict their impact on function, structural stability, antibiotic binding, and, ultimately, their effect on bacterial sensitivity to antibiotics. RESULTS Eight mutations in the rpoB-encoded protein (R421S, F450S, L456I, S454F, D461E, S476F, L478S, and S519Y) are associated with resistance to rifampin and rifalazil. These mutations occur in conserved regions of the protein, leading to decreased stability and affecting essential functional sites of RNA polymerase, the target of these antibiotics. Although the structural differences between the native and mutant proteins are minimal, notable changes in local hydrogen bonding have been observed. Despite similar binding energies, variations in hydrogen bonds and hydrophobic interactions in certain mutants (for instance, D461E for rifalazil and S476F for rifampin) indicate that these changes may diminish ligand affinity and specificity. Furthermore, protein-protein network analysis demonstrated a strong correlation between wild-type rpoB and ten C. pneumoniae proteins, each fulfilling specific functional roles. Consequently, some of these mutations can reduce the bacterium's sensitivity to rifampin and rifalazil, thereby contributing to antibiotic resistance. CONCLUSION The findings of this study indicate that mutations in the rpoB gene, which encodes the beta subunit of RNA polymerase, are pivotal in the resistance of C. pneumoniae to rifampin and rifalazil. Some of these mutations may result in reduced protein stability and changes in the structure, function, and antibiotic binding. As a consequence, the efficacy of these drugs in inhibiting RNA polymerase is compromised, allowing the bacteria to persist in transcription and replication even in the presence of antibiotics. Overall, these insights enhance our understanding of the resistance mechanisms in C. pneumoniae and could guide the development of strategies to address this challenge. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Sanae Esskhayry
- Life and Health Sciences Laboratory, Faculty of Medicine and Pharmacy, Abdelmalek Essaâdi University, Tangier, Morocco
- Department of Medical Genetics and Oncogenetics, Mohammed VI University Hospital Center, Tangier, Morocco
- Systems and Data Engineering Team, National School of Applied Sciences, Abdelmalek Essaâdi University, Tangier, 90 000, Morocco
- Chlamydiae and Mycoplasma Laboratory, Research Department, Institut Pasteur du Maroc, Casablanca, 20360, Morocco
| | - Ichrak Benamri
- Systems and Data Engineering Team, National School of Applied Sciences, Abdelmalek Essaâdi University, Tangier, 90 000, Morocco
- Laboratory of Information Technology and Modeling, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, B. P 7955 Sidi Othmane, Casablanca, Morocco
- Chlamydiae and Mycoplasma Laboratory, Research Department, Institut Pasteur du Maroc, Casablanca, 20360, Morocco
| | - Afaf Lamzouri
- Life and Health Sciences Laboratory, Faculty of Medicine and Pharmacy, Abdelmalek Essaâdi University, Tangier, Morocco
- Department of Medical Genetics and Oncogenetics, Mohammed VI University Hospital Center, Tangier, Morocco
| | - Ouafae Kaissi
- Department of Medical Genetics and Oncogenetics, Mohammed VI University Hospital Center, Tangier, Morocco
| | - Rachida Fissoune
- Systems and Data Engineering Team, National School of Applied Sciences, Abdelmalek Essaâdi University, Tangier, 90 000, Morocco
| | - Ahmed Moussa
- Systems and Data Engineering Team, National School of Applied Sciences, Abdelmalek Essaâdi University, Tangier, 90 000, Morocco
| | - Fouzia Radouani
- Chlamydiae and Mycoplasma Laboratory, Research Department, Institut Pasteur du Maroc, Casablanca, 20360, Morocco.
| |
Collapse
|
134
|
Song K, Li S, Liu J, Kang Z. Global research trend of Herpes simplex keratitis: a bibliometric analysis and visualization from 1941 to 2024. Front Med (Lausanne) 2025; 12:1526116. [PMID: 40177280 PMCID: PMC11961934 DOI: 10.3389/fmed.2025.1526116] [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: 11/27/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Objective Herpes simplex keratitis (HSK), caused by the herpes simplex virus (HSV), is a leading cause of infectious blindness worldwide. This study aims to explore the research trends, key contributors, and emerging areas of focus in HSK research through bibliometric analysis. Methods Publications related to HSK from 1941 to 2024 were retrieved from the Web of Science Core Collection (WoSCC). Bibliometric and visual analyses were conducted using VOSviewer, CiteSpace, and R 4.3.3. Results A total of 1,076 publications on HSK were identified. The top three contributing countries were the United States (267 papers), China (99), and Japan (64). Harvard University was the leading institution with 75 publications, while the American Journal of Ophthalmology emerged as the most influential journal, boasting an h-index of 29. Kaufman, HE, was the most cited author, with 1,988 citations. The top three keywords were "infection" (82), "stromal keratitis" (73), and "penetrating keratoplasty" (62). Burst keyword analysis indicated a growing interest in terms such as "outcome" and "ultraviolet A" since 2018. Conclusion This bibliometric analysis underscores two primary research areas in HSK: the clinical management of stromal keratitis and infection, as well as the mechanisms of HSK recurrence, which include strategies for preventing reactivation and managing immune rejection. Future research is anticipated to focus on innovative treatments, particularly ultraviolet A therapy.
Collapse
Affiliation(s)
| | | | | | - Zefeng Kang
- China Academy of Traditional Chinese Medicine Hospital of Ophthalmology, Beijing, China
| |
Collapse
|
135
|
Kalamvoki M. HSV-1 virions and related particles: biogenesis and implications in the infection. J Virol 2025; 99:e0107624. [PMID: 39898651 PMCID: PMC11915793 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
|
136
|
Vile R, Kendall B, Liseth O, Sangsuwannukul T, Elliott N, Yerovi MC, Thompson J, Swanson J, Rizk S, Diaz R, Tonne J. Immunodominant antiviral T cell responses outcompete immuno-subdominant antitumor responses to reduce the efficacy of oncolytic viroimmunotherapy. RESEARCH SQUARE 2025:rs.3.rs-6131273. [PMID: 40166032 PMCID: PMC11957203 DOI: 10.21203/rs.3.rs-6131273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The paradigm in the field of oncolytic virotherapy proposes that tumor cell killing by an oncolytic virus (OV) culminates in the priming of antitumor CD8 T cells. However, this ignores the impact a highly immunodominant antiviral response against the OV has on the antitumor response which has been weakened by mechanisms of central tolerance. Here, we show that inflammatory Vesicular Stomatitis Virus (VSV) failed to prime an adoptively transferred, or pre-existing, population of tumor-reactive T cells. Combination with αPD1 immune checkpoint blockade therapy improved survival only when VSV expressed tumor associated antigens (TAA). These data show that, in this model, the highly inflammatory OV VSV alone actively outcompetes antitumor immunity. However, we also show that viral expression of a mutant near-self TAA can break central tolerance expanding heteroclitic self-reactive and near-self-reactive T cells, thus overcoming viral immunodominance by promoting tumor-specific T cell proliferation in parallel with expanding antiviral T cells.
Collapse
|
137
|
Sattarnezhad N, Kockum I, Thomas OG, Liu Y, Ho PP, Barrett AK, Comanescu AI, Wijeratne TU, Utz PJ, Alfredsson L, Steinman L, Robinson WH, Olsson T, Lanz TV. Antibody reactivity against EBNA1 and GlialCAM differentiates multiple sclerosis patients from healthy controls. Proc Natl Acad Sci U S A 2025; 122:e2424986122. [PMID: 40063790 PMCID: PMC11929495 DOI: 10.1073/pnas.2424986122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 03/25/2025] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS), which is linked to Epstein-Barr virus (EBV) infection, preceding the disease. The molecular mechanisms underlying this connection are only partially understood. We previously described molecular mimicry between the EBV transcription factor EBV nuclear antigen 1 (EBNA1) and three human CNS proteins: anoctamin-2 (ANO2), alpha-B crystallin (CRYAB), and glial cellular adhesion molecule (GlialCAM). Here, we investigated antibody responses against EBNA1 and GlialCAM in a large cohort of 650 MS patients and 661 matched population controls and compared them to responses against CRYAB and ANO2. We confirmed that elevated IgG responses against EBNA1 and all three CNS-mimic antigens associate with increased MS risk. Blocking experiments confirmed the presence of cross-reactive antibodies and molecular mimicry between EBNA1 and GlialCAM, and accompanying antibody responses against adjacent peptide regions of GlialCAM suggest epitope spreading. Antibody responses against EBNA1, GlialCAM, CRYAB, and ANO2 are elevated in MS patients carrying the main risk allele HLA-DRB1*15:01, and combinations of HLA-DRB1*15:01 with anti-EBNA1 and anti-GlialCAM antibodies increase MS risk significantly and in an additive fashion. In addition, antibody reactivities against more than one EBNA1 peptide and more than one CNS-mimic increase the MS risk significantly but modestly. Overall, we show that molecular mimicry between EBNA1 and GlialCAM is likely an important molecular mechanism contributing to MS pathology.
Collapse
Affiliation(s)
- Neda Sattarnezhad
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
- Department of Neurology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94304
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm171 76, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm171 76, Sweden
| | - Olivia G. Thomas
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm171 76, Sweden
- Department of Clinical Neuroscience, Therapeutic Immune Design, Center for Molecular Medicine, Karolinska Institute, Stockholm171 77, Sweden
| | - Yicong Liu
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm171 76, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm171 76, Sweden
| | - Peggy P. Ho
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Alison K. Barrett
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Alexandros I. Comanescu
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Tilini U. Wijeratne
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Paul J. Utz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Lars Alfredsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm171 76, Sweden
- Unit of Occupational Medicine, Institute of Environmental Medicine, Karolinska Institutet, Stockholm171 77, Sweden
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Beckman Center for Molecular Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
- The Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94304
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm171 76, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm171 76, Sweden
| | - Tobias V. Lanz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
138
|
Kokudo T, Kokudo N. Impact of hepatitis C virus infection control on preventing the postoperative recurrence of intrahepatic cholangiocarcinoma. Hepatol Res 2025; 55:629-630. [PMID: 40318092 DOI: 10.1111/hepr.14182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Affiliation(s)
- Takashi Kokudo
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
139
|
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
|
140
|
Zhang Z, Wei Q, Wu C, Ye Z, Qin L, Chen T, Sun Z, Tian K, Li X. Isolation and pathogenicity of a novel recombinant pseudorabies virus from the attenuated vaccine and classical strains. Front Vet Sci 2025; 12:1579148. [PMID: 40166703 PMCID: PMC11955811 DOI: 10.3389/fvets.2025.1579148] [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: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Pseudorabies (PR) remains one of the most important swine diseases in China. Live attenuated vaccines have been widely deployed and have proven highly effective in controlling PR in the field. However, recent concerns regarding the evolution and recombination events involving pseudorabies virus (PRV) vaccine strains have raised substantial attention. In the present study, a novel recombinant PRV strain named HN2201 was isolated from one stillbirth case in Henan province in 2022. To assess the genetic and evolutionary features, the major immunogenic and virulence-associated genes, including gB, gC, gD, gG, gE and TK, were sequenced and analyzed. Phylogenetic and nucleotide homology analysis revealed that gB, gC, gD and gG genes of HN2201 displayed close relationship with Chinese classical strains. However, the TK gene of HN2201 contained a continuous deletion of 205 nucleotides, sharing the highest nucleotide homology (99.9%) with HB-98 vaccine strain. Additionally, a similar deletion was observed in the promoter region of the gE gene in both HN2201 and HB-98. Pathogenicity studies on 9-week-old piglets demonstrated that HN2201 exhibited attenuated virulence, characterized by transient clinical signs. The above results suggest that the naturally isolated HN2201 likely resulted from recombination events between the PRV classical strain and the HB-98 vaccine strain. Our findings provide valuable insights into the evolution of PRV in China and underscore the necessity of scientific and cautious use of PRV vaccines in the field.
Collapse
Affiliation(s)
- Zhendong Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Qingteng Wei
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chengyue Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhengqin Ye
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Liting Qin
- Qingdao Jiazhi Biotechnology Co. Ltd., Qingdao, China
| | - Ting Chen
- Qingdao Jiazhi Biotechnology Co. Ltd., Qingdao, China
| | - Zhe Sun
- National Research Center for Veterinary Medicine, Luoyang, China
| | - Kegong Tian
- National Research Center for Veterinary Medicine, Luoyang, China
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
141
|
Campise M. COVID-19 and Transplant Patients: Challenges, Risks, and Evolving Strategies. Vaccines (Basel) 2025; 13:318. [PMID: 40266237 PMCID: PMC11945915 DOI: 10.3390/vaccines13030318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
The first cases of COVID-19 were reported in December 2019 in Wuhan, China [...].
Collapse
Affiliation(s)
- Mariarosaria Campise
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
142
|
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
|
143
|
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
|
144
|
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] [Track Full Text] [Download PDF] [Figures] [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
|
145
|
Samoilenko M, Nedosekov V, Bertoni G. Testing the Tenacity of Small Ruminant Lentiviruses In Vitro to Assess the Potential Risk of Indirect Fomites' Transmission. Viruses 2025; 17:419. [PMID: 40143344 PMCID: PMC11946479 DOI: 10.3390/v17030419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
In 2011-2013, we isolated and characterized small ruminant lentiviruses (SRLVs) from two flocks, one of goats and the other of sheep, that had never been in direct contact. Phylogenetic analysis of these viruses indicated a common origin, which led us to hypothesize indirect transmission of these viruses between the two flocks. Since, to our knowledge, there are no published data on the tenacity of these viruses, we started this work. In the first part, we monitored the loss of infectivity of two prototypic SRLV strains, MVV 1514 and CAEV-CO, over time, in liquid suspension. As expected, the suspensions stored at 4 °C better preserved the infectivity of the viruses. Additionally, viruses resuspended in milk, the medium mirroring the in vivo situation, proved more tenacious than those maintained in a cell culture medium. These viruses, subjected to harsh treatments such as drying and resuspending, partially maintained their replication capacity. After an immediate loss of nearly 1 log10 TCID50 immediately after desiccation, the viruses maintained their replication capacity for at least three weeks when desiccated in milk. These results suggest that fomites, clothing, or pastures contaminated with secretions or milk from infected animals might mediate the infection of animals independently of direct contact.
Collapse
Affiliation(s)
- Maksym Samoilenko
- Institute of Virology and Immunology IVI, 3147 Mittelhäusern, Switzerland;
- Institute of Virology and Immunology IVI, 3012 Bern, Switzerland
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, University of Bern, 3012 Bern, Switzerland
- Department of Epizootiology, Microbiology and Virology, Faculty of Veterinary Medicine, National University of Life and Environmental Sciences of Ukraine, 03041 Kyiv, Ukraine;
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Vitalii Nedosekov
- Department of Epizootiology, Microbiology and Virology, Faculty of Veterinary Medicine, National University of Life and Environmental Sciences of Ukraine, 03041 Kyiv, Ukraine;
- Royal Veterinary College, University of London, London NW1 0TU, UK
| | - Giuseppe Bertoni
- Institute of Virology and Immunology IVI, 3147 Mittelhäusern, Switzerland;
- Institute of Virology and Immunology IVI, 3012 Bern, Switzerland
- Vetsuisse Faculty, Department of Infectious Diseases and Pathobiology, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
146
|
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] [Download PDF] [Figures] [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
|
147
|
Brandão ML, Hermsdorff HHM, Leal ACG, Bressan J, Pimenta AM. Vaccination and food consumption: association with Post-Acute COVID-19 Syndrome in Brazilian adults (CUME Study). Front Nutr 2025; 12:1549747. [PMID: 40161300 PMCID: PMC11950691 DOI: 10.3389/fnut.2025.1549747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
Background Post-Acute COVID-19 Syndrome (PACS) is an important sequalae of COVID-19. Then, our objective was to analyze the risk and protective factors for PACS in Brazilian adults participating in the Cohort of Universities of Minas Gerais (CUME Study), with emphasis on COVID-19 vaccination and food consumption. Methods In this sub-study, we included 2,065 participants of CUME Study who answered the baseline questionnaire in 2016 or 2018 or 2020 or 2022, and the follow-up COVID-19/PACS-specific questionnaire in 2023. PACS diagnosis was based on self-reporting of continuation or development of new symptoms 3 months after the initial SARS-CoV-2 infection, with these symptoms lasting for at least 2 months with no other explanation. To estimate the risk and protective factors for PACS, hierarchical multivariate statistical analysis was conducted using the Cox regression technique, producing two models: (1) focusing on consumption of macro and micronutrients; (2) focusing on consumption of food groups. Results After a median of 5.5 years of follow-up, 54.4% of the participants reported PACS. When we analyzed the consumption of macro and micronutrients, higher intake of proteins (HR: 1.36; 95% CI: 1.06-1.74-4th quartile) and lipids (HR: 1.23; 95% CI: 1.02-1.48-4th quartile) were risk factors for PACS. On the other hand, higher intake of vitamin C (HR: 0.78; 95% CI: 0.64-0.94-4th quartile), vitamin D (HR: 0.81; 95% CI: 0.67-0.99-4th quartile), and zinc (HR: 0.66; 95% CI: 0.52-0.83-4th quartile) were protective factors for the outcome (model 1). When we analyzed the consumption of food groups, higher intake of eggs (HR: 1.59; 95% CI: 1.34-1.89-4th quartile) increased the risk of PACS, whereas, respectively, higher and intermediate consumption of white meat (HR: 0.84; 95% CI: 0.71-1.00-4th quartile) and vegetables (HR: 0.81; 95% CI: 0.67-0.99-2nd quartile; HR: 0.81; 95% CI: 0.67-0.99-3rd quartile) decreased the risk of the outcome (model 2). In both models, pre-infection COVID-19 vaccination was a protective factor for PACS. Conclusion A healthy diet, with higher consumption of white meat, vegetables and specific micronutrients (vitamin C, vitamin D, zinc), in parallel with pre-infection COVID-19 vaccination, is essential to reduce the risk of PACS.
Collapse
Affiliation(s)
- Marlise Lima Brandão
- Posgraduate Program in Nursing, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | | | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | |
Collapse
|
148
|
Pashkov EA, Shikvin DA, Pashkov GA, Nagieva FG, Bogdanova EA, Bykov AS, Pashkov EP, Svitich OA, Zverev VV. Assessment of the preventive effect of knockdown of cellular genes NXF1, PRPS1 PRPS1 and NAA10 in influenza infection in an in vitro model. Vopr Virusol 2025; 70:66-77. [PMID: 40233338 DOI: 10.36233/0507-4088-289] [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] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Influenza is an acute respiratory viral infectious disease caused by the influenza viruses. Current preventive and therapeutic approaches are of great anti-epidemic importance, but there are a number of problems, such as the rapid emergence of resistant strains, the lack of cross-immunity and the effectiveness of vaccines. One of the approaches to the development of anti-influenza agents is the use of RNA interference and small interfering RNAs complementary to the mRNA target of viral and cellular genes. Aim ‒ to evaluate the prophylactic anti-influenza effect of siRNAs directed to the cellular genes NXF1, PRPS1 and NAA10 in an in vitro model. MATERIALS AND METHODS Antigenic variants of influenza A virus: A/California/7/09 (H1N1), A/WSN/33 (H1N1) and A/Brisbane/59/07 (H1N1); cell cultures A549 and MDCK. The study was performed using molecular genetic (transfection, NC isolation, RT-PCR-RV) and virological (cell culture infection, titration by visual CPE, viral titer assessment using the Ramakrishnan method) methods. RESULTS It was shown that siRNAs targeting the cellular genes NXF1, PRPS1 and NAA10, when used prophylactically in cell culture at a concentration of 0.25 μg per well, during infection with influenza virus strains A/California/7/09 (H1N1), A/WSN/33 (H1N1) and A/Brisbane/59/07 (H1N1) at a multiplicity of infection of 0.01, reduced viral replication to a level of 220 TCID50 per 1 ml of cell medium, whereas in control untreated cells the viral yield was ~106 TCID50 per 1 ml of medium. CONCLUSIONS Reproduction of influenza A viruses directly depends on the protein products of the NXF1, PRPS1, and NAA10 genes. Reduced expression of these genes disrupts the life cycle and activity of influenza viruses. Such an approach can potentially be studied and used for closely and distantly related representatives of other virus families.
Collapse
Affiliation(s)
- E A Pashkov
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - D A Shikvin
- Moscow State University of Fine Chemical Technologies
| | - G A Pashkov
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - F G Nagieva
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
| | - E A Bogdanova
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - A S Bykov
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - E P Pashkov
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - O A Svitich
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| | - V V Zverev
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
- I.I. Mechnikov Scientific and Research Institute of Vaccines and Sera
| |
Collapse
|
149
|
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
|
150
|
Zhou YQ, Jiang JX, He S, Li YQ, Cheng XX, Liu SQ, Wei PP, Guan XY, Ong CK, Wang VYF, Luo CL, Bei JX. Epstein-Barr virus hijacks histone demethylase machinery to drive epithelial malignancy progression through KDM5B upregulation. Signal Transduct Target Ther 2025; 10:83. [PMID: 40059116 PMCID: PMC11891327 DOI: 10.1038/s41392-025-02163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 05/13/2025] Open
Abstract
Epstein-Barr virus (EBV) is a significant epigenetic driver in the development of epithelial-origin nasopharyngeal carcinoma (NPC) and gastric cancer (GC), which together represent 80% of EBV-associated malignancies. Despite its known association, the specific mechanisms, particularly those involving EBV-induced histone modifications, remain poorly understood. Through integrative analyses of single-cell and bulk transcriptome data from epithelial tumor tissues and EBV-infected cells, we identified KDM5B as a critical histone-modifying factor consistently upregulated following EBV infection. We demonstrated that EBV stimulates KDM5B expression via interactions of its latent gene EBNA1 with transcription factor CEBPB and through direct binding of its lytic gene BZLF1 to Zta-response elements on the KDM5B promoter. Functional assays revealed that KDM5B acts as an oncogene, correlating with poor survival outcomes in EBV-associated epithelial cancers. Mechanistically, KDM5B inhibited the tumor suppressor gene PLK2 through histone demethylation, thereby activating the PI3K/AKT/mTOR signaling pathway and promoting malignant progression. Furthermore, treatment with the KDM5B inhibitor AS-8351 markedly attenuated this signaling activity and exhibited strong anti-tumor effect in both in vitro and in vivo patient-derived xenograft models from EBV-associated tumors. Together, these findings provide novel insights into how EBV hijacks KDM5B to mediate histone demethylation of PLK2, facilitating tumor progression through the PI3K/AKT/mTOR pathway in epithelial cancers, highlighting promising therapeutic strategies targeting epigenetic alterations in EBV-associated cancers.
Collapse
Affiliation(s)
- Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jia-Xin Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P.R. China
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Vivien Ya-Fan Wang
- Faculty of Health, University of Macau, Avenida da Universidade, Taipa, Macau SAR, P.R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore.
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, P.R. China.
- Department of Clinical Oncology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, P.R. China.
| |
Collapse
|