201
|
Kohs J, Lichtenthäler T, Gouma C, Cho HK, Reith A, Kramer A, Reiche S, Zwicker P. Studies on the Virucidal Effects of UV-C of 233 nm and 275 nm Wavelengths. Viruses 2024; 16:1904. [PMID: 39772211 PMCID: PMC11680280 DOI: 10.3390/v16121904] [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: 10/14/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Among the physical decontamination methods, treatment with ultraviolet (UV) radiation is a suitable means of preventing viral infections. Mercury vapor lamps (254 nm) used for room decontamination are potentially damaging to human skin (radiation) and harmful to the environment (mercury). Therefore, other UV-C wavelengths (100-280 nm) may be effective for virus inactivation on skin without damaging it, e.g., far-UV-C radiation with a wavelength of 233 nm, which is absorbed in the outer layer of the skin and thus does not reach the deeper layers of the skin. For room disinfection, 275 nm UV-C LED lamps could be a more environmentally friendly alternative, since toxic mercury is avoided. A carrier test using multiple viruses was used to determine the TCID50/mL value on stainless steel, PVC, and glass carriers. In addition to the inactivation kinetics (233 nm), the necessary UV-C dose for 4 lg inactivation (275 nm) was investigated. The impact of irradiance on the inactivation efficacy was also assessed. The inactivation of the viruses was a function of the radiation dose. UV-C-radiation at 233 nm (80 mJ/cm2) inactivated from 1.49 ± 0.08 to 4.28 ± 0.18 lg depending on the virus used. To achieve a 4 lg inactivation (275 nm) for enveloped viruses, doses of up to 70 mJ/cm2 (SuHV-1) were sufficient. For non-enveloped viruses, a maximum dose of 600 mJ/cm2 (MS2) was necessary. Enveloped viruses were inactivated with lower doses compared to non-enveloped viruses. Higher radiation doses were required for inactivation at 275 nm in comparison to 254 nm. A more environmentally friendly alternative to mercury vapor lamps is available with 275 nm LED emitters. Radiation at 233 nm could serve as an additional prophylactic or therapeutic measure for virus inactivation in direct contact with human skin.
Collapse
Affiliation(s)
- Jessica Kohs
- Department of Experimental Animal Facilities and Biorisk Management (ATB), Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald Insel Riems, Germany
| | - Tom Lichtenthäler
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Carolyn Gouma
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Hyun Kyong Cho
- Ferdinand-Braun-Institut gGmbH, Leibniz-Institut Für Höchstfrequenztechnik, Gustav-Kirchhoff-Str. 4, 12489 Berlin, Germany
| | - Andreas Reith
- ams OSRAM International GmbH, Leibnizstr. 4, 93055 Regensburg, Germany
| | - Axel Kramer
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sven Reiche
- Department of Experimental Animal Facilities and Biorisk Management (ATB), Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald Insel Riems, Germany
| | - Paula Zwicker
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| |
Collapse
|
202
|
Smith JC, Arunachalam PS, Legere TH, Cavacini LA, Hunter E, Pulendran B, Amara RR, Kozlowski PA. Induction of Tier 2 HIV-Neutralizing IgA Antibodies in Rhesus Macaques Vaccinated with BG505.664 SOSIP. Vaccines (Basel) 2024; 12:1386. [PMID: 39772048 PMCID: PMC11680376 DOI: 10.3390/vaccines12121386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND A goal of mucosal human immunodeficiency virus type 1 (HIV-1) vaccines is to generate mucosal plasma cells producing polymeric IgA (pIgA)-neutralizing antibodies at sites of viral entry. However, vaccine immunogens capable of eliciting IgA neutralizing antibodies (nAbs) that recognize tier 2 viral isolates have not yet been identified. METHODS To determine if stabilized native-like HIV-1 envelope (Env) trimers could generate IgA nAbs, we purified total IgA and IgG from the banked sera of six rhesus macaques that had been found in a previous study to develop serum nAbs after subcutaneous immunization with BG505.664 SOSIP and 3M-052 adjuvant, which is a TLR7/8 agonist. The neutralization of autologous tier 2 BG505 T332N pseudovirus by the IgA and IgG preparations was measured using the TZM-bl assay. Anti-SOSIP binding antibodies (bAbs) were measured by ELISA. RESULTS The IgG samples were found to have significantly greater levels of both nAb and bAb. However, after normalizing the nAb titer relative to the concentration of bAb, SOSIP-specific IgA purified from 2/6 animals was found to neutralize just as effectively as SOSIP-specific IgG, and in 3/6 animals, neutralization by the specific IgA was significantly greater. The more potent neutralization by IgA in these three animals was associated with a higher percentage of anti-SOSIP J chain-bound (polymeric) antibody. CONCLUSIONS The parenteral vaccination of nonhuman primates with BG505.664 SOSIP generates HIV-1 tier 2 IgA nAbs in serum, including SOSIP-specific polymeric IgA, which appears to neutralize more efficiently than monomeric IgA or IgG. Mucosal delivery of this SOSIP or other stable Env trimers could generate locally synthesized polymeric IgA nAbs in mucosal tissues and secretions.
Collapse
Affiliation(s)
- Justin C. Smith
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94304, USA;
| | - Traci H. Legere
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA;
| | - Lisa A. Cavacini
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA;
| | - Eric Hunter
- Emory Vaccine Center, Department of Pathology and Laboratory Medicine, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA;
| | - Bali Pulendran
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94304, USA;
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Rama R. Amara
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA;
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| |
Collapse
|
203
|
Yin H, Lin X, Gan C, Xiao H, Jiang Y, Zhou X, Yang Q, Jiang W, Wang M, Yang H, Zhang G, Chan H, Li Q. Prediction Model and Decision Analysis for Early Recognition of SDNS/FRNS in Children. J Inflamm Res 2024; 17:10585-10598. [PMID: 39670155 PMCID: PMC11635163 DOI: 10.2147/jir.s494530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose This study identified factors that identification of progression-predicting utility from steroid-sensitive nephrotic syndrome(SSNS) to steroid-dependent or frequently relapsing nephrotic syndrome (SDNS/FRNS) in patients and developed a corresponding predictive model. Patients and Methods This retrospective study analyzed clinical data from 756 patients aged 1 to 18 years, diagnosed with SSNS, who received treatment at the Department of Nephrology, Children's Hospital of Chongqing Medical University, between November 2007 and May 2023. We developed a shrinkage and selection operator (LASSO) - logistic regression model, which was visualized using a nomogram. The model's performance, validity, and clinical utility were evaluated through receiver operating characteristic curve analysis, confusion matrix, calibration plot, and decision curve analysis. Results The platelet-to-lymphocyte ratio (PLR) was identified as an independent risk factor for progression, with an odds ratio (OR) of 1.01 (95% confidence interval (CI) = 1.01-1.01, p = 0.009). Additionally, other significant factors included the time for urinary protein turned negative (OR = 1.17, 95% CI = 1.12-1.23, p < 0.001), estimated glomerular filtration rate(eGFR) (OR = 0.99, 95% CI = 0.98-0.99, p < 0.001), low-density lipoprotein (OR = 0.90, 95% CI = 0.83-0.97, p = 0.006), thrombin time (OR = 1.22, 95% CI = 1.07-1.39, p = 0.003), and neutrophil absolute counts (OR = 1.10, 95% CI = 1.02-1.18, p = 0.009). The model's performance was assessed through internal validation, yielding an area under the curve of 0.78 (0.73-0.82) for the training set and 0.81 (0.75-0.87) for the validation set. Conclusion PLR, eGFR, the time for urinary protein turned negative, low-density lipoprotein, thrombin time, and neutrophil absolute counts may be effective predictors for identifying SSNS patients at risk of progressing to SDNS/FRNS. These findings offer valuable insights for early detection and support the use of precision medicine strategies in managing SDNS/FRNS.
Collapse
Affiliation(s)
- Hui Yin
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Xiao Lin
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Chun Gan
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Han Xiao
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Yaru Jiang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Xindi Zhou
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Qing Yang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Wei Jiang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Mo Wang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Haiping Yang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Gaofu Zhang
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Han Chan
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| | - Qiu Li
- Department of Nephrology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, People’s Republic of China
| |
Collapse
|
204
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. Sci Immunol 2024; 9:eadp7951. [PMID: 39642242 DOI: 10.1126/sciimmunol.adp7951] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/08/2024] [Indexed: 12/08/2024]
Abstract
Severity of COVID-19 is affected by multiple factors; however, it is not understood how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure affects the control of viral replication. Here, we demonstrate that immune events in the mouse lung closely preceding SARS-CoV-2 infection affect viral control and identify innate immune pathways that limit viral replication. Pulmonary inflammatory stimuli including resolved, antecedent respiratory infections with Staphylococcus aureus or influenza, ongoing pulmonary Mycobacterium tuberculosis infection, ovalbumin/alum-induced asthma, or airway administration of TLR ligands and recombinant cytokines all establish an antiviral state in the lung that restricts SARS-CoV-2 replication. In addition to antiviral type I interferons, TNFα and IL-1 potently precondition the lung for enhanced viral control. Our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation preceding SARS-CoV-2 exposure may contribute to variability in disease outcomes.
Collapse
Affiliation(s)
- Paul J Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Andrea C Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Eduardo P Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sydnee T Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Artur T L Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jaspal S Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, MD 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, MD 20852, USA
| | - Daniel L Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bruno B Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
205
|
Tian Y, Dai J, Yang Y, Guo X, Wang W, Li F, Wang J, Liu R. Relationship between the risk of intestinal mucosal Epstein-Barr virus and/or cytomegalovirus infection and peripheral blood NK cells numbers in patients with ulcerative colitis: a cross-sectional study in Chinese population. Front Microbiol 2024; 15:1498483. [PMID: 39697654 PMCID: PMC11652489 DOI: 10.3389/fmicb.2024.1498483] [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: 09/19/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Objective This study aimed to analyze the relationship between the risk of common opportunistic pathogens Epstein-Barr virus (EBV) and cytomegalovirus (CMV) infection in intestinal mucosal tissues of Ulcerative Colitis (UC) patients and the number of peripheral blood NK cells. Methods UC patients admitted to a third-grade class-A hospital from January 2018 to December 2023 were selected as research population. Clinical data of the patients were collected from the electronic medical record system. Additionally, samples of intestinal mucosal tissues were obtained for real-time fluorescence quantitative PCR to detect and analyze the viral load of CMV and EBV. Blood samples were collected for lymphocyte subsets analysis. Multivariable logistic regression models analyses was used to determine the odds ratio (OR) and 95% confidence interval (95% CI) for the independent association between NK cells and EBV/CMV infections in UC. We further applied the restricted cubic spline analysis and smooth curve fitting to examine the non-linear relationship between them. Results 378 UC patients were enrolled. Of these patients, there were 194 patients (51.32%) with EBV /CMV infection. In multivariable logistic regression analyses NK cells was independently associated with EBV and/or CMV infection after adjusted potential confounders (OR 8.24, 95% CI 3.75-18.13, p < 0.001). A nonlinear relationship was found between NK cells and EBV/CMV infections, which had a threshold around 10.169. The effect sizes and CIs below and above the threshold were 0.535 (0.413-0.692), p < 0.001 and 1.034 (0.904-1.183), p > 0.05, respectively. Conclusion There was a non-linear relationship between NK cells and EBV/CMV infections. The risk for EBV/CMV infections was not increased with increasing NK cells in individuals with NK cells ≥ 10.169%, whereas the risk for EBV and/or CMV infection was increased with an decreasing NK cells in those with NK cells < 10.169%. The risk of EBV/CMV infections increases when NK cells were below a certain level.
Collapse
Affiliation(s)
- Ye Tian
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Jinghua Dai
- School of Nursing, Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Yunfeng Yang
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Xiaofeng Guo
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Wei Wang
- Department of Laboratory Medicine, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Fengxia Li
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, National Clinical Research Center for Digestive Diseases, Shanxi Inflammatory Bowel Disease Center, Taiyuan, China
| | - Juzi Wang
- Nursing Department, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Ruiyun Liu
- Shanxi Children’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
206
|
Gedefaw A, Tadesse BT, Tadesse S, Kebede B, Hussen S, Hailu D, Berhan Y, Makonnen E, Vella S, Aklillu E. The progress of mother-to-child transmission of Human Immunodeficiency Virus (HIV) after Dolutegravir (DTG) optimization program: evidence from a multicenter cohort study in Ethiopia. BMC Public Health 2024; 24:3367. [PMID: 39627710 PMCID: PMC11613881 DOI: 10.1186/s12889-024-20761-w] [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: 05/01/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Ethiopia aims to eliminate mother-to-child transmission (MTCT) of HIV by 2030. In 2020, Dolutegravir-based antiretroviral treatment (ART) regimen optimization was done for the Prevention of Mother-to-Child Transmission (PMTCT). However, data tracking progress, particularly post-rollout of the Dolutegravir (DTG)-based regimen, and the real-world effectiveness of the new regimen are unavailable. METHODS A multicenter retrospective cohort study was conducted among HIV-infected mothers and their HIV-exposed infants visiting the selected hospitals for routine care. Eligible participants were HIV-exposed infants enrolled in the PMTCT care from 2017 to 2022. However, only the 2021 and 2022 birth cohorts were considered post-DTG optimization considering 2020 a year of optimization. The cumulative incidence of perinatal MTCT tested at 6-8 weeks of infant age, and end of care MTCT tested at 18 months of age was assessed. The exposures of the study were the infant birth cohort years and the different ART regimens used for PMTCT of HIV. RESULTS Among a total of 2,643 routine care enrolled participants, 2521 (95.4%) HIV-exposed infants were included in the analysis. Of these, 210 were on follow-up and excluded from the breastfeeding MTCT analysis. A total of 30/2521(1.2%) [95% confidence interval (CI): 0.8-1.7%] were positive for HIV at 6-8 weeks. Additionally, 11 /2281 (0.50%) (95% CI: 0.3-0.9%) were positive during breastfeeding. At the end of the care, 41/2311 (1.8%) (95% CI: 1.3-2.4%) infants were HIV-positive. The highest end-of-care MTCT was reported in 2019 and 2022 birth cohorts while the lowest was in 2018 (P-value > 0.3). However, after adjusting for baseline characteristics, the trend showed a decrease in transmission rates following the rollout of DTG-based regimen, although statistical significance was not reached. The adjusted odds ratios (AORs) for perinatal, breastfeeding, and end-of-care transmission rates were 0.34 (95%CI: 0.08-1.39), 0.29(95%CI: 0.03-3.05), and 0.38(95%CI: 0.11-1.26) respectively. Compared with the Efavirenz (EFV)-based regimen, the DTG-based regimen was associated with a lower risk of MTCT in both the perinatal (AOR 0.23, 95% CI: 0.06-0.85) and at the end of care (AOR 0.27, 95% CI: 0.09-0.82). Pregnant women who started ART at late gestation had the highest transmission rate regardless of ART regimens (P-value < 0.001). CONCLUSIONS In the studied cohort population, we observed less than 3% MTCT rate at the end of PMTCT care. The findings might suggest the achievement of MTCT elimination at the hospital level. Although the DTG-based regimen demonstrated a lower risk of transmission, other contributing factors, such as late ART initiation, should be urgently addressed. Future research should focus on prospective designs, interventions targeting late ART initiation, and understanding regional disparities to further advance efforts to eliminate MTCT by 2030.
Collapse
Affiliation(s)
- Abel Gedefaw
- College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia.
- Department of Global Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Birkneh Tilahun Tadesse
- College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
- Department of Global Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sintayehu Tadesse
- PREGART clinical trial project, Hawassa University, Hawassa, Ethiopia
| | - Biruk Kebede
- PREGART clinical trial project, Hawassa University, Hawassa, Ethiopia
| | - Siraj Hussen
- College of Medicine and Health Sciences, Department of Medical Laboratory Science, Hawassa University, Hawassa, Ethiopia
| | - Dejene Hailu
- School of Public Health, Hawassa University, Hawassa, Ethiopia
| | - Yifru Berhan
- St. Paul's Millennium Medical College and Hospital, Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Center for Innovative Drug Development for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Stefano Vella
- Department of Global Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eleni Aklillu
- Department of Global Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
207
|
Bellini T, Fueri E, Formigoni C, Mariani M, Villa G, Finetti M, Marin M, De Chiara E, Bratta A, Vanorio B, Casabona F, Pepino C, Castagnola E, Piccotti E, Moscatelli A. Usefulness of Point-of-Care Testing for Respiratory Viruses in a Pediatric Emergency Department Setting. J Clin Med 2024; 13:7368. [PMID: 39685826 DOI: 10.3390/jcm13237368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Respiratory tract infections (RTIs) are a leading cause of pediatric emergency department (PED) visits, especially in children under five. These infections are primarily viral, complicating diagnosis and management. This study assesses the impact of point-of-care (POC) rapid diagnostic tests for respiratory viruses on clinical and economic outcomes in a PED setting. Materials and Methods: A retrospective analysis of 1396 POC tests for RSV, adenovirus, and influenza A/B was conducted in the PED of the Giannina Gaslini Institute, Genoa, Italy, from December 2022 to April 2024. Demographics, blood tests, admissions, and readmission rates were evaluated. Statistical analyses were performed using appropriate tests for categorical and continuous variables. Results: Of the tests, 31.5% were positive for at least one virus. Positive patients were younger and had higher hospitalization rates (50.7% vs. 39.9%) but fewer blood tests (38.2% vs. 51.7%). Economic analysis indicated lower costs for virus-positive patients. RSV-positive patients showed a significant association with higher hospitalization rates (67.8%) and readmission within 72 h. Conclusions: POC testing significantly improves patient management in PEDs by enabling rapid diagnoses, reducing unnecessary tests and hospitalizations, and guiding appropriate treatment. This approach supports better resource allocation, crucial during peak seasons, and has implications for reducing antibiotic use and resistance. Further research is warranted to explore long-term impacts on patient outcomes and healthcare efficiency.
Collapse
Affiliation(s)
- Tommaso Bellini
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Elena Fueri
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Clelia Formigoni
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16147 Genoa, Italy
| | - Marcello Mariani
- Infectious Diseases Unit and COVID Hospital, Department of Pediatrics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Giovanna Villa
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Martina Finetti
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Marta Marin
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Elena De Chiara
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16147 Genoa, Italy
| | - Anna Bratta
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16147 Genoa, Italy
| | - Barbara Vanorio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16147 Genoa, Italy
| | - Federica Casabona
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16147 Genoa, Italy
| | - Carlotta Pepino
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Elio Castagnola
- Infectious Diseases Unit and COVID Hospital, Department of Pediatrics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Emanuela Piccotti
- Paediatric Emergency Room and Emergency Medicine Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, via G. Gaslini, 5, 16147 Genoa, Italy
| | - Andrea Moscatelli
- Pediatric and Neonatal Intensive Care Unit, Emergency Department, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
208
|
Barkyoumb D, Tavakol SA, Zhao X, Stephens ME, Bageac DV, Bowen IE, Desai VR. Povidone-iodine-induced scalp lesions in pediatric neurosurgery patients: a case series. Childs Nerv Syst 2024; 41:27. [PMID: 39623026 DOI: 10.1007/s00381-024-06677-1] [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: 09/09/2024] [Accepted: 10/29/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE Povidone-iodine, or Betadine® (Atlantis Consumer Healthcare Inc., Bridgewater, NJ), is a commonly used agent for surgical site preparation. Although commonly used, it carries the risk of skin reactions, and multiple cases of intra-operative contact dermatitis and chemical burns have been reported. However, to our knowledge, there are currently no published cases of povidone-iodine-induced skin lesions in neurosurgical patients. METHODS A single-center retrospective chart review was performed to identify patients who developed scalp lesions secondary to chemical toxicity following neurosurgical procedures between October 1, 2021, and June 30, 2024. RESULTS Three patients were identified, ranging from 2.5 months to 14 years old. Two were positioned prone and the third in lateral decubitus. All patients' heads were supported by a horseshoe headrest covered with a gel pad and wrapped in a cotton roll. For two patients, Reston™ foam (3M©, St. Paul, MN) was added on the horseshoe. Surgical sites were prepped with isopropyl alcohol, Betadine®, and chlorohexidine. Two patients had their heads intermittently lifted throughout the procedure. Lesions were identified immediately after returning the patient to the supine position in the operating room and steadily improved over the course of one to five months with local wound care. CONCLUSIONS Careful preparation of the surgical site is an essential step in the prevention of surgical site infections. However, caution should be exercised during skin preparation to avoid pooling of povidone-iodine around dependent regions. Additional steps, such as scheduled head elevations and pressure dispersion, should be taken to mitigate factors contributing to these lesions.
Collapse
Affiliation(s)
- David Barkyoumb
- Department of Neurosurgery, Section of Pediatric Neurosurgery, Oklahoma Children's Hospital, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sherwin A Tavakol
- Department of Neurosurgery, Section of Pediatric Neurosurgery, Oklahoma Children's Hospital, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiaochun Zhao
- Department of Neurosurgery, Section of Pediatric Neurosurgery, Oklahoma Children's Hospital, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mark E Stephens
- Department of Neurosurgery, Section of Pediatric Neurosurgery, Oklahoma Children's Hospital, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Devin V Bageac
- Division of Neurosurgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Neurosurgery, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Ira E Bowen
- Department of Neurosurgery, Section of Pediatric Neurosurgery, Oklahoma Children's Hospital, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Virendra R Desai
- Division of Neurosurgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Neurosurgery, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
209
|
Harrache A, Saker K, Mokdad B, Generenaz L, Saade C, Pons S, Fassier JB, Bal A, Trabaud MA, Rabilloud M, Abichou-Klich A, Trouillet-Assant S. Anti-RBD IgG dynamics following infection or vaccination. Vaccine 2024; 42:126464. [PMID: 39432992 DOI: 10.1016/j.vaccine.2024.126464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/10/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Identifying parameters influencing SARS-CoV-2 antibody dynamics post infection or vaccination is crucial for refining vaccination strategies. In a longitudinal analysis of 1340 samples from 375 healthcare workers, we characterized peak serological response and IgG half-life. Peak antibody titers post 2 vaccine doses were ∼ 20 times higher than natural infection; conversely, infected individuals had extended antibody half-life. Clinical and demographical factors such as BMI, age and smoking shaped peak response without affecting anti-RBD IgG half-life. A third mRNA vaccine dose increased peak antibody titers and prolonged half-life compared to the second dose. These findings underscore the diverse kinetics of SARS-CoV-2 antibody responses, which is influenced by immunization type/number and clinical factors.
Collapse
Affiliation(s)
- Amira Harrache
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Kahina Saker
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Bouchra Mokdad
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Laurence Generenaz
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Carla Saade
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Sylvie Pons
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| | - Jean-Baptiste Fassier
- Occupational Health and Medicine Department, Hospices Civils de Lyon, 69002 Lyon, France
| | - Antonin Bal
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Mary-Anne Trabaud
- Virology Laboratory, Institute of Infectious Agents, Laboratory associated with the National Reference Centre for Respiratory Infection Viruses, Hospices Civils de Lyon, IAI, North Biology Centre, North Hospital Group, 69004 Lyon, France
| | - Muriel Rabilloud
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Amna Abichou-Klich
- Biostatistics Department, Hospices Civils de Lyon, Lyon France. CNRS, UMR 5558, University of Lyon, Biometrics and Evolutionary Biology Laboratory, Biostatistics-Health Team, 69100 Villeurbanne, France
| | - Sophie Trouillet-Assant
- Joint Research Unit Hospices Civils of Lyon-bioMérieux, Lyon Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France.
| |
Collapse
|
210
|
Mohd Hisham AA, Mat Yassim AS, Suppian R, Azlan M, Mohamad Asri AA, Idris NS, Muhamad R, Norazmi MN. Comparable and sustained levels of S1-RBD-IgG and S1-RBD-IgA in BNT162b2 homologous and CoronaVac-BNT162b2 heterologous booster vaccination: A 22-month prospective study in Malaysia. Vaccine 2024; 42:126471. [PMID: 39490114 DOI: 10.1016/j.vaccine.2024.126471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
This prospective cohort study examines the long-term humoral immune responses post-COVID-19 vaccination in 146 individuals who received either a homologous three-dose BNT162b2 vaccine regimen (PPP) or two primary doses of CoronaVac followed by BNT162b2 booster (SSP) in Malaysia. The study focuses on serum anti-S1-RBD-IgG, -IgA, and -IgM, using the ELISA method. The results show that BNT162b2 outperformed CoronaVac in the two dose primary vaccination series. BNT162b2 booster dose significantly raised serum anti-S1-RBD-IgG and -IgA levels, sustaining this increase from 26 to 52 weeks after administration, regardless of the vaccine regimen. This leads to equivalent levels of anti-S1-RBD-IgG and -IgA after boosting with BNT162b2 in both groups. Breakthrough infections, particularly with the emergence of the Omicron variant, did not result in increased anti-S1-RBD-IgG and -IgA levels. No significant induction of anti-S1-RBD-IgM was observed following multiple vaccine doses. The long-term investigation revealed that PPP and SSP groups had comparable humoral immune responses to SARS-CoV-2, highlighting the advantage of mRNA booster dose in our cohort.
Collapse
Affiliation(s)
- Anis Atifah Mohd Hisham
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Aini Syahida Mat Yassim
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia.
| | - Rapeah Suppian
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Maryam Azlan
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | | | - Nur Suhaila Idris
- School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rosediani Muhamad
- School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia; Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000 Kajang, Selangor, Malaysia.
| |
Collapse
|
211
|
Chatterjee S, Tilley H, Briordy D, Waldron RT, Kordbacheh R, Cutts WD, Cook A, Pandol SJ, Kim BJ, Fairweather D, Sin J. Investigating the potential role of capsaicin in facilitating the spread of coxsackievirus B3 via extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626352. [PMID: 39677700 PMCID: PMC11642798 DOI: 10.1101/2024.12.02.626352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Coxsackievirus B3 (CVB3) is a non-enveloped picornavirus that can cause systemic inflammatory diseases including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. We have previously reported that following infection, CVB3 localizes to mitochondria, inducing mitochondrial fission and mitophagy, while inhibiting lysosomal degradation by blocking autophagosome-lysosome fusion. This results in the release of virus-laden mitophagosomes from the host cell as infectious extracellular vesicles (EVs) which allow non-lytic viral egress. Transient receptor potential vanilloid 1 (TRPV1/ TRPV1 ) is a heat and capsaicin-sensitive cation channel that regulates mitochondrial dynamics by inducing mitochondrial membrane depolarization and fission. In this study, we found that treating cells with the TRPV1 agonist capsaicin dramatically enhances CVB3 egress via EVs. Analysis of the released EVs revealed increased levels of viral capsid protein VP1/ VP1 , mitochondrial protein TOM70/ TOMM70 , and fission protein phospho-DRP1/ DNM1L (Ser 616). Moreover, these EVs exhibited increased levels of heat shock protein HSP70/ HSPA1A , suggesting a potential role of these chaperones in facilitating infectious EV release from cells. Furthermore, TRPV1 inhibition with capsazepine significantly reduced viral infection in vitro . We previously observed similar effects in vitro with another TRPV1 inhibitor SB-366791. Our current in vivo studies found that SB-366791 significantly mitigates pancreatic damage and reduces viral titers in mouse model of CVB3 pancreatitis. Given the lack of understanding regarding the factors that contribute to diverse clinical manifestations of CVB3, our study highlights capsaicin and TRPV1 as potential exacerbating factors that facilitates CVB3 dissemination via mitophagy-derived EVs. IMPORTANCE CVB3 is a prevalent pathogen responsible for a range of severe diseases, including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. Despite its clinical significance, factors that determine the severity of CVB3 infection and why some individuals experience life-threatening manifestations while others have mild, cold-like symptoms remain poorly understood. This study provides new insights into the molecular mechanisms underlying CVB3 dissemination and pathogenesis. By investigating the role of capsaicin, a common dietary component, in modulating viral spread, we demonstrate that activation of TRPV1 by capsaicin enhances release of infectious CVB3 via mitophagy-derived EVs. Our results offer novel evidence that modulating TRPV1 activity could influence the clinical outcomes of CVB3 infection, opening new avenues for therapeutic interventions. Given the widespread consumption of capsaicin, this study highlights an important dietary factor that could play a role in shaping CVB3 pathogenesis and its clinical manifestations, underscoring the potential for targeted strategies to mitigate severe disease outcomes.
Collapse
|
212
|
Gu SX, Marcus BS, Gu VW, Varghese AP, Hwa J, Faustino EVS. High-Dimensional Single-Cell Mass Cytometry Demonstrates Differential Platelet Functional Phenotypes in Infants With Congenital Heart Disease. Arterioscler Thromb Vasc Biol 2024; 44:2530-2539. [PMID: 39171400 PMCID: PMC11602369 DOI: 10.1161/atvbaha.124.321131] [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: 04/21/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Congenital heart disease (CHD) is a group of complex heart defects associated with hematologic abnormalities, including increased risk of thrombotic and bleeding events. Past studies have observed evidence of platelet hyperreactivity, while other studies showed decreased platelet activation in patients with CHD. The goal of this study was to develop a mass spectrometry approach to characterize single platelets in infants with CHD and identify potential etiology for such discrepant results. METHODS We enrolled 19 infants with CHD along with 21 non-CHD controls at Yale New Haven Children's Heart Center. A single-cell high-dimensional mass cytometry method was developed to quantitatively interrogate platelet surface markers in whole blood. Additionally, plasma cytokine analysis was performed through a multiplexed panel of 52 vascular and inflammatory markers to assess for platelet releasates. RESULTS We found that infants with CHD had significant differences in platelet activation and functional markers by mass cytometry compared with non-CHD controls. Based on cell surface markers, we classified the platelets into 8 subpopulations (P0 to P7). Distinct subpopulations of platelets (P1, P4, and P5) exhibiting decreased aggregatory phenotype but altered secretory phenotypes were also identified and found to be more abundant in the blood of infants with CHD. Electron microscopy identified increased proportion of hypogranular platelets in CHD. Moreover, cytokine analysis demonstrated an overall increase in plasma cytokines and biomarkers in CHD, including IL (interleukin)-6, IL-8, IL-27, RANTES (regulated upon activation, normal T cell expressed and secreted), and VWF (von Willebrand factor), which are expressed in platelet granules and can be released upon activation. CONCLUSIONS We developed a robust mass cytometry approach to identify platelet phenotypic heterogeneity. Infants with CHD had alterations in distinct subpopulations of platelets with overall reduced aggregatory phenotype and secretory dysfunction. These findings suggest that platelets in infants with CHD may be exhausted due to persistent stimulation and may explain both bleeding and thrombotic vascular complications associated with CHD.
Collapse
Affiliation(s)
- Sean X. Gu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT
| | - Brian S. Marcus
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT
| | - Vivian W. Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Adarsh P. Varghese
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
213
|
Aryaloka S, Khairullah AR, Kusala MKJ, Fauziah I, Hidayatik N, Agil M, Yuliani MGA, Novianti AN, Moses IB, Purnama MTE, Wibowo S, Fauzia KA, Raissa R, Furqoni AH, Awwanah M, Riwu KHP. Navigating monkeypox: identifying risks and implementing solutions. Open Vet J 2024; 14:3144-3163. [PMID: 39927376 PMCID: PMC11799651 DOI: 10.5455/ovj.2024.v14.i12.1] [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: 08/05/2024] [Accepted: 11/02/2024] [Indexed: 02/11/2025] Open
Abstract
Monkeypox is a zoonotic disease caused by the orthopox virus, a double-stranded DNA virus that belongs the Poxviridae virus family. It is known to infect both animals (especially monkeys and rodents) and humans and causes a rash similar to smallpox. Humans can become infected with monkeypox virus (MPXV) when they get in close contact with infected animals (zoonotic transmission) or other infected people (human-human transmission) through their body fluids such as mucus, saliva, or even skin sores. Frequently observed symptoms of this disease include fever, headaches, muscle aches, and a rash that initially looks like a tiny bump before becoming a lump that is filled with fluid. Monkeypox symptoms also include an incubation period of 5-21 days, divided into prodromal and eruption phases. Several contributing factors, such as smallpox vaccine discontinuation, widespread intake of infected animal products as a source of protein, and high population density, amongst others, have been linked to an increase in the frequency of monkeypox outbreaks. The best course of action for diagnosing individuals who may be suffering from active monkeypox is to collect a sample of skin from the lesion and perform PCR molecular testing. Monkeypox does not presently have a specific therapy; however, supportive care can assist in managing symptoms, such as medication to lower body temperature and pain. Three major orthopoxvirus vaccines have been approved to serve as a preventive measure against monkeypox: LC16, JYNNEOS, and ACAM2000. The discovery that the monkeypox outbreak is communicable both among humans and within a population has sparked new public health worries on the possibility of the outbreak of another viral pandemic. Research and studies are still being conducted to gain a deeper understanding of this zoonotic viral disease. This review is therefore focused on deciphering monkeypox, its etiology, pathogenesis, transmission, risk factors, and control.
Collapse
Affiliation(s)
- Suhita Aryaloka
- Master Program of Veterinary Agribusiness, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | | | - Ima Fauziah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Nanik Hidayatik
- Division of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Agil
- Division of Veterinary Clinic Reproduction and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - M. Gandul Atik Yuliani
- Division of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Arindita Niatazya Novianti
- Division of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ikechukwu Benjamin Moses
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Syahputra Wibowo
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Kartika Afrida Fauzia
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| | - Ricadonna Raissa
- Department of Pharmacology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Abdul Hadi Furqoni
- Center for Biomedical Research, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Mo Awwanah
- Research Center for Applied Botany, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Katty Hendriana Priscilia Riwu
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Pendidikan Mandalika, Mataram, Indonesia
| |
Collapse
|
214
|
Xu S, Tang Y, Li M, Zhang L, Su Y, Wang Y, Liu Y, Shen Y. Clinical characteristics of Chinese children with EV-D68-associated pneumonia: A single-center retrospective analysis. Pediatr Pulmonol 2024; 59:3660-3666. [PMID: 39315747 DOI: 10.1002/ppul.27285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/19/2024] [Accepted: 09/14/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Human enterovirus D68 (EV-D68) has been associated with an increase in mild-to-severe pediatric respiratory diseases in western countries. However, the prevalence and clinical characteristics of EV-D68-associated pneumonia in China remain understudied. METHODS Between January 2022 and January 2024, 28 patients with EV-D68-associated pneumonia were enrolled. We described the prevalence, demographic, and clinical characteristics of patients with EV-D68-associated pneumonia. RESULTS Among the 28 enrolled patients, the male-to-female ratio was 1.5:1, and the average age at onset was 4.6 ± 2.7 years. Four (14.3%) required intensive care support. Monoinfection occurred in 11 cases (39.3%), while coinfections were seen in 17 cases (60.7%). 82.1% of patients had a history of one or more atopic diseases. The primary symptoms of EV-D68-associated pneumonia included cough (100%), wheezing (53.6%), and fever (53.6%). Radiologically, patchy opacity was the predominant feature, observed in 72.7% of cases. No statistically significant differences were found in symptoms, laboratory tests, or imaging findings between the monoinfection and coinfection groups. Except for one case who developed quadriplegia sequelae, all patients had a favorable prognosis. CONCLUSION EV-D68 is not a common pathogen for community-acquired pneumonia in China. It mainly affects young children, particularly those with atopic constitution. The overall prognosis is favorable, although neurological complications are rare and may lead to severe sequelae. This study is the first investigation into the prevalence and clinical characteristics of EV-D68-associated pneumonia in China.
Collapse
Affiliation(s)
- Shasha Xu
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yu Tang
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Min Li
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Lei Zhang
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yanyan Su
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yanqiong Wang
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yuemei Liu
- Respiratory Department, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yuelin Shen
- Respiratory Department II, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
215
|
Dinh MTP, Iqbal M, Abhishek K, Lam FW, Shevkoplyas SS. Recent developments in microfluidic passive separation to enable purification of platelets for transfusion. BIOMICROFLUIDICS 2024; 18:061504. [PMID: 39713738 PMCID: PMC11658822 DOI: 10.1063/5.0226060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Platelet transfusion is a lifesaving therapy intended to prevent and treat bleeding. However, in addition to platelets, a typical unit also contains a large volume of supernatant that accumulates multiple pro-inflammatory contaminants, including residual leukocytes, microaggregates, microparticles, antibodies, and cytokines. Infusion of this supernatant is responsible for virtually all adverse reactions to platelet transfusions. Conventional methods for removing residual leukocytes (leukoreduction) and reducing the volume of transfused supernatant (volume reduction) struggle to mitigate these risks holistically. Leukoreduction filters can remove leukocytes and microaggregates but fail to reduce supernatant volume, whereas centrifugation can reduce volume, but it is ineffective against larger contaminants and damages platelets. Additionally, platelet purification based on these methods is often too logistically complex, time-consuming, and labor-intensive to implement routinely. Emerging microfluidic technologies offer promising alternatives through passive separation mechanisms that enable cell separation with minimal damage and drastically reduced instrumentation size and facility requirements. This review examines recent innovations in microfluidic cell separation that can be used for leukoreduction and volume reduction of platelets. It begins by defining the performance requirements that any separation method must meet to successfully replace conventional methods currently used to perform these tasks. Standard performance metrics are described, including leukocyte depletion efficiency, degree of volume reduction, processing throughput, and platelet recovery. Finally, the review outlines the primary challenges that must be overcome to enable simple-to-use, disposable microfluidic devices capable of both reducing the platelet unit volume and removing pro-inflammatory contaminants, while preserving most functional platelets for transfusion.
Collapse
Affiliation(s)
- Mai T. P. Dinh
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, USA
| | - Mubasher Iqbal
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, USA
| | - Kumar Abhishek
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, USA
| | - Fong W. Lam
- Division of Pediatric Critical Care Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Sergey S. Shevkoplyas
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77204, USA
| |
Collapse
|
216
|
Fischli K, Schöbi N, Duppenthaler A, Casaulta C, Riedel T, Kopp MV, Agyeman PKA, Aebi C. Postpandemic fluctuations of regional respiratory syncytial virus hospitalization epidemiology: potential impact on an immunization program in Switzerland. Eur J Pediatr 2024; 183:5149-5161. [PMID: 39331153 PMCID: PMC11527947 DOI: 10.1007/s00431-024-05785-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
RSV hospitalization epidemiology is subject to rapid changes brought about by the COVID-19 pandemic and the prospect of vaccine prevention. The purpose of this report is to characterize recent epidemiologic and clinical fluctuations and to analyze their potential impact on an immunization program with nirsevimab. This is a 2018-2024 retrospective analysis of all hospitalizations caused by RSV in patients below 16 years of age occurring at an academic Children's Hospital that serves a defined population. We simulated the vaccine impact against RSV hospitalization by applying the expected effects of the infant immunization program with nirsevimab proposed in Switzerland to observed case counts. We analyzed 1339 hospitalizations. The consecutive occurrence of two major epidemics in 2022-2023 and 2023-2024 had never been recorded previously. The 2023-2024 season witnessed a major shift to older age. Only 61% of patients were below 12 months of age, while prepandemic long-term surveillance since 1997 found a range between 64 and 85% (median, 73%). Age below 3 months, prematurity, airway anomalies, congenital heart disease, and neuromuscular disorders were independently associated with ICU admission. Simulation of the vaccine impact using two scenarios of coverage and efficacy (scenario 1, 50% and 62%, respectively; scenario 2, 90% and 90%) and three different age distributions resulted in an infant vaccine impact of 31.0% (scenario 1) and 81.0% (scenario 2), respectively. Vaccine impact for all patients below 16 years ranged from 22.7 to 24.9% (scenario 1) and 54.2 to 68.8% (scenario 2). CONCLUSION RSV hospitalization epidemiology was characterized by substantial variability in patient age on admission. As the proposed RSV immunization program primarily targets infants, year-to-year fluctuation of cases among older children will cause a variability of vaccine impact of approximately 15%. This information may be useful for physicians and hospital administrators when they anticipate the resources needed during the winter season. WHAT IS KNOWN • RSV hospitalization epidemiology was subject to massive disturbances during the COVID-19 pandemic. • Extended half-life monoclonal antibodies and active maternal immunization offer new means of passive protection of infants against severe RSV disease. WHAT IS NEW • We demonstrate substantial year-to-year fluctuation of the age distribution at the time of RSV hospitalization. • Up to 40% of annual RSV hospitalizations in a given season occur in children above 12 months of age who do not benefit from maternal RSV immunization and may not be eligible for receipt of a monoclonal antibody.
Collapse
Affiliation(s)
- Klara Fischli
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland
| | - Nina Schöbi
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland
| | - Andrea Duppenthaler
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland
| | - Carmen Casaulta
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Thomas Riedel
- Division of Pediatric Intensive Care Medicine, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Matthias V Kopp
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland
- Airway Research Center North (ARCN), Member of the German Lung Research Center (DZL), University of Lübeck, Lübeck, Germany
| | - Philipp K A Agyeman
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland
| | - Christoph Aebi
- Division of Pediatric Infectious Disease, Department of Pediatrics, Bern University Hospital, Inselspital, University of Bern, CH-3010, Bern, Switzerland.
| |
Collapse
|
217
|
Hafner L, Gadin E, Huang L, Frouin A, Laporte F, Gaultier C, Vieira A, Maudet C, Varet H, Moura A, Bracq-Dieye H, Tessaud-Rita N, Maury M, Dazas M, Legendre R, Gastineau P, Tsai YH, Coppée JY, Charlier C, Patin E, Chikhi R, Rocha EPC, Leclercq A, Disson O, Aschard H, Lecuit M. Differential stress responsiveness determines intraspecies virulence heterogeneity and host adaptation in Listeria monocytogenes. Nat Microbiol 2024; 9:3345-3361. [PMID: 39578578 DOI: 10.1038/s41564-024-01859-8] [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: 03/21/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
Microbial pathogenesis is mediated by the expression of virulence genes. However, as microbes with identical virulence gene content can differ in their pathogenic potential, other virulence determinants must be involved. Here, by combining comparative genomics and transcriptomics of a large collection of isolates of the model pathogen Listeria monocytogenes, time-lapse microscopy, in vitro evolution and in vivo experiments, we show that the individual stress responsiveness of L. monocytogenes isolates determines their respective levels of virulence in vivo and reflects their degree of host adaptation. The transcriptional signature that accounts for the heterogeneity in the virulence of L. monocytogenes species is mediated by the stress response regulator SigB and driven by differential stress responsiveness. The tuning of SigB pathway responsiveness is polygenic and influenced by multiple, individually rare gene variations. This study reveals an overarching determinant of microbial virulence, challenging the paradigm of accessory virulence gene content as the major determinant of intraspecies virulence heterogeneity.
Collapse
Affiliation(s)
- Lukas Hafner
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Enzo Gadin
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Lei Huang
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Arthur Frouin
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Fabien Laporte
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Charlotte Gaultier
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Afonso Vieira
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Claire Maudet
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Hugo Varet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alexandra Moura
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Hélène Bracq-Dieye
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Nathalie Tessaud-Rita
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Mylène Maury
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Melody Dazas
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Rachel Legendre
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pauline Gastineau
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Yu-Huan Tsai
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Jean-Yves Coppée
- Transcriptome et Epigenome Platform, Biomics, Center for Technological Resources and Research, Institut Pasteur, Université Paris Cité, Paris, France
| | - Caroline Charlier
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
- Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, Institut Imagine, APHP, Paris, France
| | - Etienne Patin
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
| | - Rayan Chikhi
- Sequence Bioinformatics Group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Eduardo P C Rocha
- Microbial Evolutionary Genomics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3525, Paris, France
| | - Alexandre Leclercq
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France
| | - Olivier Disson
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France
| | - Hugues Aschard
- Statistical Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS USR375, Paris, France
| | - Marc Lecuit
- Biology of Infection Unit, Institut Pasteur, Université Paris Cité, Inserm U1117, Paris, France.
- National Reference Center and WHO Collaborating Center Listeria, Institut Pasteur, Paris, France.
- Necker-Enfants Malades University Hospital, Division of Infectious Diseases and Tropical Medicine, Institut Imagine, APHP, Paris, France.
| |
Collapse
|
218
|
Marcos-Villar L, Perdiguero B, López-Bravo M, Zamora C, Sin L, Álvarez E, Sorzano CÓS, Sánchez-Cordón PJ, Casasnovas JM, Astorgano D, García-Arriaza J, Anthiya S, Borrajo ML, Lou G, Cuesta B, Franceschini L, Gelpí JL, Thielemans K, Sisteré-Oró M, Meyerhans A, García F, Esteban I, López-Bigas N, Plana M, Alonso MJ, Esteban M, Gómez CE. Heterologous mRNA/MVA delivering trimeric-RBD as effective vaccination regimen against SARS-CoV-2: COVARNA Consortium. Emerg Microbes Infect 2024; 13:2387906. [PMID: 39087555 PMCID: PMC11313003 DOI: 10.1080/22221751.2024.2387906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
Despite the high efficiency of current SARS-CoV-2 mRNA vaccines in reducing COVID-19 morbidity and mortality, waning immunity and the emergence of resistant variants underscore the need for novel vaccination strategies. This study explores a heterologous mRNA/Modified Vaccinia virus Ankara (MVA) prime/boost regimen employing a trimeric form of the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein compared to a homologous MVA/MVA regimen. In C57BL/6 mice, the RBD was delivered during priming via an mRNA vector encapsulated in nanoemulsions (NE) or lipid nanoparticles (LNP), followed by a booster with a replication-deficient MVA-based recombinant virus (MVA-RBD). This heterologous mRNA/MVA regimen elicited strong anti-RBD binding and neutralizing antibodies (BAbs and NAbs) against both the ancestral SARS-CoV-2 strain and different variants of concern (VoCs). Additionally, this protocol induced robust and polyfunctional RBD-specific CD4 and CD8 T cell responses, particularly in animals primed with mLNP-RBD. In K18-hACE2 transgenic mice, the LNP-RBD/MVA combination provided complete protection from morbidity and mortality following a live SARS-CoV-2 challenge compared with the partial protection observed with mNE-RBD/MVA or MVA/MVA regimens. Although the mNE-RBD/MVA regimen only protects half of the animals, it was able to induce antibodies with Fc-mediated effector functions besides NAbs. Moreover, viral replication and viral load in the respiratory tract were markedly reduced and decreased pro-inflammatory cytokine levels were observed. These results support the efficacy of heterologous mRNA/MVA vaccine combinations over homologous MVA/MVA regimen, using alternative nanocarriers that circumvent intellectual property restrictions of current mRNA vaccine formulations.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Mice, Inbred C57BL
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Humans
- Female
- Nanoparticles/administration & dosage
- Vaccination
- mRNA Vaccines/administration & dosage
- Mice, Transgenic
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- CD8-Positive T-Lymphocytes/immunology
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Liposomes
Collapse
Affiliation(s)
- Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Carmen Zamora
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | - Pedro J. Sánchez-Cordón
- Veterinary Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
| | | | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mireya L. Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Gustavo Lou
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Belén Cuesta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Lorenzo Franceschini
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Josep L. Gelpí
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona (UB), Barcelona, Spain
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marta Sisteré-Oró
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department, Hospital Clínic, UB,Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Ignasi Esteban
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - Núria López-Bigas
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Madrid, Spain
| | - Montserrat Plana
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, UB, Barcelona, Spain
| | - María J. Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigaciones Sanitarias (IDIS), Santiago de Compostela, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
219
|
Šušol O, Šušolová B, Klempíř O, Navrátil M, Gumulec J, Kořístek Z, Ďuraš J, Kaščák M, Mihályová J, Stejskal L, Jelínek T, Richterová P, Szeligová L, Plonková H, Zuchnická J, Dluhošová B, Demel I, Buffa D, Hradská K, Popková T, Muroňová L, Lachnit M, Lančová K, Hájek R. One Year Duration of Immune Response Following a 3rd Booster Dose of mRNA Vaccine Against COVID-19 in 292 Patients With Hematological Malignancies in University Hospital Ostrava, Czech Republic. Cancer Med 2024; 13:e70503. [PMID: 39711119 DOI: 10.1002/cam4.70503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024] Open
Abstract
AIMS To evaluate antibody response to mRNA vaccine, identify subgroups with poor response and to determine long-term antibody durability in hematological patients. MATERIALS AND METHODS We have vaccinated 292 patients with all hematological malignancies with a third dose of mRNA COMIRNATY vaccine with a 12-month follow-up period in our center in Ostrava, Czech Republic. RESULTS Antibody response for the whole cohort exceeded 74% through the whole 12-month follow-up. Lowest seroconversion was observed in CLL cohort (20/41, 48.8%), patients who received anti-CD20 therapy < 6 months before vaccination (8/30, 26.7%) and BTK inhibitors (3/6, 50.0%). On the contrary, patients with chronic myeloproliferative neoplasms and acute leukemia performed comparably with healthy population (33/33; 100% and 12/13; 92.3%, respectively). We have seen better results if the time interval between anti-CD20 therapy and additional vaccine dose was longer than 6 months (5/8 patients achieved seroconversion on 4th booster dose after previous failure). Also, 36 patients received a 4th dose of vaccine as a booster with measurable increase in protective antibodies in 50% (18/36). CONCLUSIONS Additional doses show promise for a well-timed revaccination even in poor responders. To our knowledge, no study comparable to our work in terms of follow-up length, vaccine consistency or variety of hematological malignancies and/or treatment has been reported yet. Our findings shed more light on long-term antibody response to mRNA vaccines against SARS-CoV-2 in patients with hematological cancer and bring important data for the evaluation of possible vaccine failure and scheduling of subsequent doses.
Collapse
Affiliation(s)
- Ondrej Šušol
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Barbora Šušolová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Ondřej Klempíř
- Department of Biomedical Informatics, Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Milan Navrátil
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jaromír Gumulec
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zdeněk Kořístek
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Juraj Ďuraš
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Michal Kaščák
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Jana Mihályová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Lukáš Stejskal
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematology and Blood Transfusion, Silesian Hospital in Opava, Opava, Czech Republic
| | - Tomáš Jelínek
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Petra Richterová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Lenka Szeligová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Hana Plonková
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Jana Zuchnická
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Barbora Dluhošová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ivo Demel
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - David Buffa
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Katarína Hradská
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tereza Popková
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ludmila Muroňová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Martin Lachnit
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Klára Lančová
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haemato-Oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
220
|
Ott C, Dutilh G, Reist J, Bingisser R, Egli A, Heininger U. Clinical Presentation of Enterovirus D68 in a Swiss Pediatric University Center. Pediatr Infect Dis J 2024; 43:1135-1140. [PMID: 39163309 PMCID: PMC11542972 DOI: 10.1097/inf.0000000000004503] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Enterovirus D68 (EV-D68) is responsible for millions of infections. In the last decade, there has been an increase in the number of children requiring hospital or critical care admission due to severe respiratory illness. Nevertheless, the epidemiological and clinical importance of EV-D68 infections remains unclear. OBJECTIVE We aimed to determine the local prevalence of EV-D68 infection in pediatric patients and to characterize its clinical presentation and disease burden compared with non-EV-D68 enterovirus and human rhinovirus (RV) infections. STUDY DESIGN We performed a retrospective single-center study of children presenting with respiratory symptoms and positive respiratory panel polymerase chain reaction for EV/RV from November 2018 to December 2019. We tested EV/RV positive specimens with an EV-D68-specific polymerase chain reaction to discriminate EV-D68, non-EV-D68 and RV and compared their respective clinical presentation, outcomes and treatment. RESULTS We identified 224 patients (median age 21 months), of which 16 (7%) were EV-D68 positive. They presented with cough (88%), wheezing (62%) and dyspnea (75%). EV-D68 infection had an odds ratio regarding pediatric respiratory severity-score of 11.6 relative to non-EV-D68 [confidence intervals (CI): 3.51-41.14], and of 9.9 (CI: 3.75-27.95) relative to RV. The fitted logistic regression showed that the odds of intensive care were 5 times more likely with EV-D68 than RV infection (CI: 1.32-19.28; P = 0.001). Patients with EV-D68 infections were more likely to receive medical support in the form of supplementary oxygen, antibiotics and steroids. CONCLUSIONS EV-D68 infection is associated with higher morbidity and a higher likelihood of intensive care treatment than non-EV-D68 and RV infections.
Collapse
Affiliation(s)
- Chantal Ott
- From the Applied Microbiology Research, Department of Biomedicine, University of Basel
- Department of Pediatric infectious diseases, Children University Hospital Basel
| | - Gilles Dutilh
- Department of Clinical Research, University of Basel
| | - Josiane Reist
- From the Applied Microbiology Research, Department of Biomedicine, University of Basel
| | | | - Adrian Egli
- From the Applied Microbiology Research, Department of Biomedicine, University of Basel
- Department of Clinical Bacteriology and Microbiology, University Hospital Basel, Basel
- Institute for Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Ulrich Heininger
- Department of Pediatric infectious diseases, Children University Hospital Basel
| |
Collapse
|
221
|
Terstappen J, Hak SF, Bhan A, Bogaert D, Bont LJ, Buchholz UJ, Clark AD, Cohen C, Dagan R, Feikin DR, Graham BS, Gupta A, Haldar P, Jalang'o R, Karron RA, Kragten L, Li Y, Löwensteyn YN, Munywoki PK, Njogu R, Osterhaus A, Pollard AJ, Nazario LR, Sande C, Satav AR, Srikantiah P, Stein RT, Thacker N, Thomas R, Bayona MT, Mazur NI. The respiratory syncytial virus vaccine and monoclonal antibody landscape: the road to global access. THE LANCET. INFECTIOUS DISEASES 2024; 24:e747-e761. [PMID: 39326422 DOI: 10.1016/s1473-3099(24)00455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 09/28/2024]
Abstract
Respiratory syncytial virus (RSV) is the second most common pathogen causing infant mortality. Additionally, RSV is a major cause of morbidity and mortality in older adults (age ≥60 years) similar to influenza. A protein-based maternal vaccine and monoclonal antibody (mAb) are now market-approved to protect infants, while an mRNA and two protein-based vaccines are approved for older adults. First-year experience protecting infants with nirsevimab in high-income countries shows a major public health benefit. It is expected that the RSV vaccine landscape will continue to develop in the coming years to protect all people globally. The vaccine and mAb landscape remain active with 30 candidates in clinical development using four approaches: protein-based, live-attenuated and chimeric vector, mRNA, and mAbs. Candidates in late-phase trials aim to protect young infants using mAbs, older infants and toddlers with live-attenuated vaccines, and children and adults using protein-based and mRNA vaccines. This Review provides an overview of RSV vaccines highlighting different target populations, antigens, and trial results. As RSV vaccines have not yet reached low-income and middle-income countries, we outline urgent next steps to minimise the vaccine delay.
Collapse
Affiliation(s)
- Jonne Terstappen
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sarah F Hak
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anant Bhan
- Yenepoya Medical College & Centre for Ethics, Yenepoya University, Mangalore, India
| | - Debby Bogaert
- Paediatric Medicine, University of Edinburgh, Edinburgh, UK
| | - Louis J Bont
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands; ReSViNET Foundation, Zeist, Netherlands
| | - Ursula J Buchholz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew D Clark
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Cheryl Cohen
- Center for Respiratory Diseases and Meningitis, University of the Witwatersrand and National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Ron Dagan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheba, Israel
| | - Daniel R Feikin
- Department of Immunization, Vaccines, and Biologicals, WHO, Geneva, Switzerland
| | - Barney S Graham
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Anuradha Gupta
- Global Immunization, Sabin Vaccine Institute, Washington, DC, USA
| | - Pradeep Haldar
- Government of India, Ministry of Health and Family Welfare, Delhi, India
| | - Rose Jalang'o
- National Vaccines and Immunization Program, Ministry of Health, Nairobi, Kenya
| | - Ruth A Karron
- Boomberg School of Public Health Department of International Health, Johns Hopkins Bloomberg Baltimore, MD, USA
| | | | - You Li
- Centre for Global Health, University of Edinburgh, Edinburgh, UK; School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yvette N Löwensteyn
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Rosemary Njogu
- Department of International Health, Jhpiego, Nairobi, Kenya
| | - Ab Osterhaus
- Center of Infection Medicine and Zoonosis Research, University of Veterinary Medicine, Hannover, Germany
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK
| | | | - Charles Sande
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ashish R Satav
- MAHAN Trust Mahatma Gandhi Tribal Hospital, Melghat, India
| | - Padmini Srikantiah
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Renato T Stein
- Pneumologia Pediátrica, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Naveen Thacker
- International Pediatric Association, Webster Groves, MI, USA; Child Health Foundation, Mumbai, India
| | | | | | - Natalie I Mazur
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
222
|
Förster C, Nordhoff K, Fritzemeier J, Freise F, Kreienbrock L. Informative Value of a Sample Investigation with a Predefined Sample Size using the Example of Listeria monocytogenes in Food Safety. J Food Prot 2024; 87:100388. [PMID: 39490987 DOI: 10.1016/j.jfp.2024.100388] [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: 01/29/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Foodborne diseases, especially those caused by zoonotic agents, pose a significant threat to human health. Food business operators are therefore responsible for producing safe food. To do this, they must regularly carry out appropriate sample investigations to detect zoonotic agents in their food before it leaves the factory. Depending on the issue investigated, there may be legal requirements for food business operators, usually specifying minimum sample sizes, such as Commission Regulation (EU) No 2073/2005. However, in most cases, there is no information on the precision, and therefore, on the significance of the results for these sample sizes. Regulatory veterinary authorities have a control function and, as a result, they are required to regularly evaluate the available investigations and their results. In addition, in certain crisis situations (e.g., foodborne outbreaks or suspected food contamination), authorities may guide food business operators in their investigations and conduct their own investigations to assess food safety measures. In such cases, as there are no legally defined sample sizes to be taken, the appropriate sample sizes must be determined by the authorities. This can lead to a conflict between the need for feasibility and the need for conclusiveness of the investigation potentially being a challenge for the regulatory authority in charge. This paper highlights the importance of thoughtful study design and the critical communication of available results by veterinary authorities on the background of a use case involving Listeria monocytogenes findings in a crisis situation. Using the minimum sample size, n = 5, required by Commission Regulation (EU) No 2073/2005 for the pathogen Listeria monocytogenes as a guide, the uncertainties associated with small sample sizes are highlighted. It also aims to facilitate the evaluation of studies performed and the assessment of further sample sizes.
Collapse
Affiliation(s)
- Cara Förster
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health in the Human-Animal-Environment Interface, University for Veterinary Medicine, Bünteweg 2, 30559 Hannover, Germany.
| | - Katja Nordhoff
- Task Force Consumer Protection, Lower Saxony State Office for Consumer Protection and Food Safety, Stau 75, 26122 Oldenburg, Germany.
| | - Jörg Fritzemeier
- Veterinary Service Osnabrück, Am Schölerberg 1, 49082 Osnabrück, Germany.
| | - Fritjof Freise
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health in the Human-Animal-Environment Interface, University for Veterinary Medicine, Bünteweg 2, 30559 Hannover, Germany.
| | - Lothar Kreienbrock
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health in the Human-Animal-Environment Interface, University for Veterinary Medicine, Bünteweg 2, 30559 Hannover, Germany.
| |
Collapse
|
223
|
Yaugel-Novoa M, Noailly B, Jospin F, Pizzorno A, Traversier A, Pozzetto B, Waeckel L, Longet S, Pillet S, Botelho-Nevers E, Rosa-Calatrava M, Bourlet T, Paul S. Impaired mucosal IgA response in patients with severe COVID-19. Emerg Microbes Infect 2024; 13:2401940. [PMID: 39358866 PMCID: PMC11451292 DOI: 10.1080/22221751.2024.2401940] [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: 06/12/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Several studies have investigated the antibody response to SARS-CoV-2, focusing particularly on the systemic humoral immune response and the production of immunoglobulin G (IgG) antibodies. IgA antibodies play a crucial role in protecting against respiratory viral infections but have also been associated with the pathophysiology of COVID-19. We performed a prospective study of 169 COVID-19 patients - 50 with critical/severe (ICU), 47 with moderate (Non-ICU), and 72 with asymptomatic COVID-19 - to explore the humoral immune response to SARS-CoV-2 infection. We found that the early systemic IgA response strongly induced in patients with severe disease did not block IgG neutralization functions and activated FcRs more effectively than IgG. However, even if SIgA levels were high, mucosal IgA antibodies could not control the infection effectively in patients with severe disease. Our findings highlight the complexity of the immune response to SARS-CoV-2 exhibiting high systemic levels of IgA with strong neutralizing capacity in severe cases, together with higher levels of IgA-FcR activation than in asymptomatic patients. They also suggest the need for further research to fully understand the role of IgA and its structural alterations in mucosal tissues in cases of severe disease and the impact of these antibodies on disease progression.
Collapse
Affiliation(s)
- Melyssa Yaugel-Novoa
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Blandine Noailly
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Fabienne Jospin
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Andrés Pizzorno
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aurélien Traversier
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Bruno Pozzetto
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Louis Waeckel
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphanie Longet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
| | - Sylvie Pillet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Diseases Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Manuel Rosa-Calatrava
- Team VirPath, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, CIRI - Centre International de Recherche en Infectiologie, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Thomas Bourlet
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Infectious Agents and Hygiene Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR530, CIC 1408 Vaccinology, CIRI – Centre International de Recherche en Infectiologie, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Lead contact
| |
Collapse
|
224
|
Doté J, Joffret ML, Beta BN, Ait-Ahmed M, Banga-Mingo V, Knowles NJ, Jouvenet N, MBaïkoua MN, Gouandjika-Vasilache I, Bessaud M. Characterization of enteroviruses circulating among farm animals and children in Central African Republic. Emerg Microbes Infect 2024; 13:2368212. [PMID: 38864685 PMCID: PMC11212570 DOI: 10.1080/22221751.2024.2368212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/10/2024] [Indexed: 06/13/2024]
Abstract
To characterize enteroviruses (EVs) circulating in farm animals in Central African Republic (CAR), we screened 192 stools of animals under 12 months belonging to family farms located in or near Bangui. To assess whether EV exchanges exist between these animals and humans, we also screened 195 stools of children who lived in contact with farm animals, as well as control stools of 358 children with no contact with farm animals. EVs were typed based on their capsid sequences.In children, all EVs belonged to species A, B and C, with EV-Cs accounting for 60%. Some EV-Cs shared recent common ancestors with lineages of vaccine-derived poliovirus that emerged in the country in 2019-2020. In animals, we identified EV-Gs that belonged to 10 different types, including a previously unknown one that we named EV-G28, while no EV-E or EV-F were observed. The CAR EV-Gs were genetically closely related to specimens sampled in other continents and some of them harboured the torovirus-derived insertion already reported in some EV-Gs. The worldwide circulation of EV-Gs is likely due the massive international trade of live animals. Besides, two human EV-Cs (coxsackievirus A17 and coxsackievirus A24) were detected in pigs, suggesting that these viruses could cross the species barrier. Our work provides original data on the epidemiology and ecology of EVs circulating among herd animals in Africa.
Collapse
Affiliation(s)
- Joël Doté
- Institut Pasteur de Bangui, Laboratoire des virus entériques/rougeole, Bangui, Central African Republic
| | - Marie-Line Joffret
- Institut Pasteur, Université de Paris Cité, Virus Sensing and Signaling Unit, Paris, France
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Paris, France
| | - Bertille Ndombari Beta
- Institut Pasteur de Bangui, Laboratoire des virus entériques/rougeole, Bangui, Central African Republic
| | - Mohand Ait-Ahmed
- Institut Pasteur, Université de Paris Cité, Pôle de coordination de la Recherche clinique, Paris, France
| | - Virginie Banga-Mingo
- Institut Pasteur de Bangui, Laboratoire des virus entériques/rougeole, Bangui, Central African Republic
| | | | - Nolwenn Jouvenet
- Institut Pasteur, Université de Paris Cité, Virus Sensing and Signaling Unit, Paris, France
| | | | | | - Maël Bessaud
- Institut Pasteur, Université de Paris Cité, Virus Sensing and Signaling Unit, Paris, France
- Laboratoire associé au Centre national de référence entérovirus/paréchovirus, Paris, France
| |
Collapse
|
225
|
Yang J, Xiao L, Zhang L, Luo G, Ma Y, Wang X, Zhang Y. Platelets: A Potential Factor that Offers Strategies for Promoting Bone Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:631-643. [PMID: 38482796 DOI: 10.1089/ten.teb.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Bone defects represent a prevalent category of clinical injuries, causing significant pain and escalating health care burdens. Effectively addressing bone defects is thus of paramount importance. Platelets, formed from megakaryocyte lysis, have emerged as pivotal players in bone tissue repair, inflammatory responses, and angiogenesis. Their intracellular storage of various growth factors, cytokines, and membrane protein receptors contributes to these crucial functions. This article provides a comprehensive overview of platelets' roles in hematoma structure, inflammatory responses, and angiogenesis throughout the process of fracture healing. Beyond their application in conjunction with artificial bone substitute materials for treating bone defects, we propose the potential future use of anticoagulants such as heparin in combination with these materials to regulate platelet number and function, thereby promoting bone healing. Ultimately, we contemplate whether manipulating platelet function to modulate bone healing could offer innovative ideas and directions for the clinical treatment of bone defects. Impact statement Given that 5-10% of fracture patients with delayed bone healing or even bone nonunion, this review explores the potential role of platelets in bone healing (directly/indirectly) and proposes ideas and directions for the future as to whether it is possible to promote bone healing and improve fracture healing rates by modulating platelets.
Collapse
Affiliation(s)
- Jingjing Yang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Lijia Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Guochen Luo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| |
Collapse
|
226
|
Uwimbabazi JC, Mutesa L, Mennechet FJ, Muvunyi CM, Kabanyana JF, Habimana RM, Mazarati JB, Mukagatare I, Iragena JDD, Moussaoui KE, Melin P, Hayette MP, Bontems S. Diversity, geographical distribution and predictive factors of Hepatitis C virus genotypes and subtypes in Rwanda. Acta Trop 2024; 260:107433. [PMID: 39447954 DOI: 10.1016/j.actatropica.2024.107433] [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: 06/07/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Existing data on the prevalence of hepatitis C virus (HCV) genotypes and subtypes in Rwanda need to be strengthened. The aim of this study was to identify HCV genotypes and subtypes among HCV-infected patients, as well as their geographical distribution in Rwanda, and to identify the social and economic factors that could influence HCV epidemiology which would make it possible to target national preventive and management actions for infected patients. METHODS This study included 560 patients with confirmed chronic HCV infection. Patients were recruited from various health facilities in the four provinces of Rwanda as well as in the City of Kigali and had never received treatment with direct-acting antiviral (DAAs). HCV viral loads were measured using Cobas® AmpliPrep/Cobas® TaqMan® HCV Quantitative Test, version 2.0. HCV genotyping was performed using an in-house sequencing protocol targeting the NS5B central region. Genotypic HCV prevalence was correlated with patient geographic location, sociodemographic, behavioral, lifestyle, and clinical factors. RESULTS HCV genotype 4 was detected in 99.3% of the patients, while genotype 3 was identified in 0.7%. A total of eight (8) HCV subtypes were detected, with 4k being the predominant subtype nationwide (49.5%), followed by subtypes 4r (21.2%), 4q (16.2%), 4v (7.9%), 4b (2.0%), 4l (1.8%), 4c and 3h represent 0.7% each. Our findings reveal subtype distribution variations among provinces. Subtype 4k was prevalent across regions, particularly in Kigali (64.0%) and the Eastern Province (61.6%). Subtype 4q was more common in the northern province (40.7%), 4r in the southern (43.9%) and western provinces (37.1%), and 4v in the eastern province (17.8%). Farmers exhibit a distinct infection profile compared to other occupations, showing a lower prevalence of subtype 4k but a higher prevalence of subtype 4r. CONCLUSIONS Our study revealed that HCV infection is unevenly distributed in Rwanda, dominated by HCV genotype 4, with considerable heterogeneity in the repartition of the different subtypes. We found potential associations between rural/urban lifestyles and HCV subtype profiles. Determined HCV distribution and diversity can serve as basis not only for HCV infection awareness and prevention campaigns, but also success and guidance for personalized treatment.
Collapse
Affiliation(s)
| | - Léon Mutesa
- Center for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda.
| | - Franck Jd Mennechet
- Pathogenesis and Control of Chronic and Emerging Infections (PCCEI) U1058, University of Montpellier, Montpellier, France
| | | | | | | | | | | | - Jean de Dieu Iragena
- Department of HIV, TB, Hepatitis and Sexually Transmitted Infections, World Health Organization/AFRO, Brazzaville, Congo
| | - Khalid El Moussaoui
- Department of Clinical Microbiology, CHU of Liege, University of Liege - Liege, Belgium
| | - Pierrette Melin
- Department of Clinical Microbiology, CHU of Liege, University of Liege - Liege, Belgium
| | - Marie-Pierre Hayette
- Department of Clinical Microbiology, CHU of Liege, University of Liege - Liege, Belgium
| | - Sébastien Bontems
- Department of Clinical Microbiology, CHU of Liege, University of Liege - Liege, Belgium.
| |
Collapse
|
227
|
Huang D, Tran L, Li JYH, Lee W, Kim E, Moussa K. Povidone iodine-infused pars plana vitrectomy for severe Arthrographis kalrae fungal endophthalmitis. Am J Ophthalmol Case Rep 2024; 36:102207. [PMID: 39553913 PMCID: PMC11566710 DOI: 10.1016/j.ajoc.2024.102207] [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: 04/18/2024] [Revised: 08/22/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Arthrographis kalrae is an opportunistic fungus that can cause a severe and atypical endophthalmitis. In limited studies, low-dose povidone iodine has been shown to be effective in treating fulminant bacterial endophthalmitis. In this case report, we describe the successful treatment of recalcitrant Arthrographis kalrae keratitis-associated endophthalmitis with intraocular povidone iodine during pars plana vitrectomy. Observations A patient with Arthrographis kalrae keratitis-associated endophthalmitis had persistent disease despite aggressive medical management. The infection resolved following pars plana vitrectomy with infusion of dilute povidone iodine. No toxicity was noted postoperatively. Conclusions and importance Povidone iodine-infused pars plana vitrectomy offers promise as a treatment for infectious endophthalmitis that fails medical management. This is the first report demonstrating eradication of fungal endophthalmitis using povidone iodine-infused pars plana vitrectomy.
Collapse
Affiliation(s)
- Denis Huang
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Lillian Tran
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Jennifer YH. Li
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - William Lee
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Esther Kim
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Kareem Moussa
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
228
|
Mahmoud YH, Eysa B, Ahmed EMS, Abdelaziz H, Zayed AM, Baki AA, Hosny A, Hassany M. Effect of treatment of chronic hepatitis c virus patients with direct-acting anti-retroviral drugs on semen and hormonal parameters. Clin Exp Reprod Med 2024; 51:309-313. [PMID: 38853129 PMCID: PMC11617913 DOI: 10.5653/cerm.2023.06772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/30/2024] [Accepted: 02/22/2024] [Indexed: 06/11/2024] Open
Abstract
OBJECTIVE Hepatitis C virus (HCV) infection is known to influence the seminal and hormonal parameters of infected men. This study was performed to assess the effects of HCV clearance using direct-acting antiviral (DAA) agents on semen and hormonal parameters. METHODS A total of 50 patients with chronic HCV were enrolled, and conventional semen analysis was performed according to World Health Organization guidelines. Basal levels of total testosterone, free testosterone (FT), follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2), prolactin, and sex hormone-binding globulin (SHBG) were assessed before and 3 months after treatment with DAAs. RESULTS Following DAA treatment, statistically significant increases were observed in sperm motility and the proportion of grade A sperm. Additionally, the percentage of abnormal forms was significantly decreased after treatment (p=0.000). However, no significant differences were observed in semen volume, concentration, or total sperm count. Sex hormone analysis of patients after DAA treatment revealed significant increases in FT, LH, and FSH levels, along with significant decreases in SHBG, prolactin, and E2 levels. CONCLUSION Following HCV clearance, we noted an improvement in sperm motility and an increase in the percentage of sperm with normal morphology. Treatment with DAAs was also associated with increased levels of FT and LH, along with decreased levels of SHBG, prolactin, and E2.
Collapse
Affiliation(s)
- Yosra H. Mahmoud
- Department of Clinical Pathology, Gastrointerology and Infectious Diseases, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Basem Eysa
- Department of Hepatology, Gastrointerology and Infectious Diseases, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Eman Mohamed Salah Ahmed
- Department of Dermatology, Andrology, Sexual Medicine and STDS, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Heba Abdelaziz
- Public Health Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Ashgan Mohamed Zayed
- Department of Medical Microbiology, Faculty of Medicine, Port-Said University, Port Said, Egypt
| | - Amin Abdel Baki
- Department of Hepatology, Gastrointerology and Infectious Diseases, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Ahmed Hosny
- Department of Dermatology, Andrology, Sexual Medicine and STDS, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Mohamed Hassany
- Tropical Medicine Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| |
Collapse
|
229
|
Coutant F, Touret F, Pin JJ, Alonzo M, Baronti C, Munier S, Attia M, de Lamballerie X, Ferry T, Miossec P. Neutralizing and enhancing monoclonal antibodies in SARS-CoV-2 convalescent patients: lessons from early variant infection and impact on shaping emerging variants. Emerg Microbes Infect 2024; 13:2307510. [PMID: 38240255 PMCID: PMC10829827 DOI: 10.1080/22221751.2024.2307510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Serological studies of COVID-19 convalescent patients have identified polyclonal lineage-specific and cross-reactive antibodies (Abs), with varying effector functions against virus variants. Individual specificities of anti-SARS-CoV-2 Abs and their impact on infectivity by other variants have been little investigated to date. Here, we dissected at a monoclonal level neutralizing and enhancing Abs elicited by early variants and how they affect infectivity of emerging variants. B cells from 13 convalescent patients originally infected by D614G or Alpha variants were immortalized to isolate 445 naturally-produced anti-SARS-CoV-2 Abs. Monoclonal antibodies (mAbs) were tested for their abilities to impact the cytopathic effect of D614G, Delta, and Omicron (BA.1) variants. Ninety-eight exhibited robust neutralization against at least one of the three variant types, while 309 showed minimal or no impact on infectivity. Thirty-eight mAbs enhanced infectivity of SARS-CoV-2. Infection with D614G/Alpha variants generated variant-specific (65 neutralizing Abs, 35 enhancing Abs) and cross-reactive (18 neutralizing Abs, 3 enhancing Abs) mAbs. Interestingly, among the neutralizing mAbs with cross-reactivity restricted to two of the three variants tested, none demonstrated specific neutralization of the Delta and Omicron variants. In contrast, cross-reactive mAbs enhancing infectivity (n = 3) were found exclusively specific to Delta and Omicron variants. Notably, two mAbs that amplified in vitro the cytopathic effect of the Delta variant also exhibited neutralization against Omicron. These findings shed light on functional diversity of cross-reactive Abs generated during SARS-CoV-2 infection and illustrate how the balance between neutralizing and enhancing Abs facilitate variant emergence.
Collapse
Affiliation(s)
- Frédéric Coutant
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
- Immunology Department, Lyon-Sud Hospital, Hospices Civils of Lyon, Pierre-Bénite, France
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Jean-Jacques Pin
- Eurobio Scientific/Dendritics – Edouard Herriot Hospital, Lyon, France
| | - Marina Alonzo
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
| | - Cécile Baronti
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Sandie Munier
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Molecular Genetics of RNA Viruses Unit, Paris, France
| | - Mikaël Attia
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Molecular Genetics of RNA Viruses Unit, Paris, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Tristan Ferry
- Department of Infectious and Tropical Diseases, Hospices Civils of Lyon - Croix-Rousse Hospital, Lyon, France
- CIRI, Inserm U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
- Department of Immunology and Rheumatology, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
230
|
Espeleta-Fox A, García-Salido A, Vallespín-Casas A, Leoz-Gordillo I, Unzueta-Roch JL, De Lama Caro-Patón G, García-Teresa MÁ, Martínez de Azagra-Garde A. Impact of nirsevimab on admission to a Spanish pediatric intensive care unit because of RSV bronchiolitis: Unicentric observational study from 2017 to 2024. Pediatr Pulmonol 2024; 59:3783-3786. [PMID: 39193993 DOI: 10.1002/ppul.27231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/29/2024]
Affiliation(s)
- Ana Espeleta-Fox
- Pediatric Critical Care Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Alberto García-Salido
- Pediatric Intensive Care Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | | - Inés Leoz-Gordillo
- Pediatric Critical Care Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | | | | | | | |
Collapse
|
231
|
Sellers RS, Ramaiah L, Hong SJ, Nambiar P, Jacquinet E, Naidu S. Session 4: mRNA and Self-Amplifying RNA (saRNA): Opportunities for Disease Prevention and Therapy. Toxicol Pathol 2024; 52:545-552. [PMID: 39578668 DOI: 10.1177/01926233241298572] [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: 11/24/2024]
Abstract
The unprecedented speed of developing vaccines against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the COVID-19 pandemic, has propelled mRNA technologies into the public eye. The versatility of mRNA technology, often referred to as "plug and play," offers immense promise for rapidly updating vaccines to address newer variants of respiratory diseases and combat emerging infectious diseases and lethal pathogens, such as the Ebolavirus. However, the potential applications of mRNA technology extend well beyond prophylactic vaccines. This session explored the two primary mRNA platforms: nonreplicating mRNA and self-amplifying mRNA (variably referred to as saRNA, samRNA, or SAM). Presentation topics were on current research efforts aimed at broadening the applications of mRNA modalities beyond vaccines. Topics included opportunities for delivering mRNA via intra-tumoral and inhalational routes, immunological and systemic inflammatory responses elicited by these modalities, and regulatory considerations involved in the development and licensing of these technologies.
Collapse
Affiliation(s)
- Rani S Sellers
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lila Ramaiah
- Johnson and Johnson, New Brunswick, New Jersey, USA
| | | | | | | | - Shan Naidu
- Moderna, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
232
|
Deniz Y, Demircioğlu B, Kuter-Emeklioğlu İ, Ekmen S, Sevınc E, Dogan E. Rotavirus Infections: A 2-Year Comprehensive Review in Admitted Pediastric Patients Amid Conflicting National Policies. Foodborne Pathog Dis 2024; 21:738-744. [PMID: 39185568 DOI: 10.1089/fpd.2024.0019] [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: 08/27/2024] Open
Abstract
Highlighting rotavirus (RV) as a significant food and waterborne pathogen, particularly affecting infants and children, causing serious gastrointestinal infections and dehydration, is important. It should be noted that there are significant debates regarding the effectiveness and outcomes of RV vaccination. In contrast to Turkey's nonmandatory vaccination policy, many developed countries implement mandatory vaccination policies, raising questions about their impact on disease prevalence and healthcare expenditures. Our study aims to comprehensively understand RV infections in Turkey and compare them with countries that have mandatory vaccination policies. We found similar, and even better, hospitalization rates, length of hospital stays, and laboratory parameters demonstrating the effectiveness of Turkey's independent vaccination approach. These findings contribute valuable insights to global vaccination strategies and disease control.
Collapse
Affiliation(s)
- Yusuf Deniz
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - Burak Demircioğlu
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - İrem Kuter-Emeklioğlu
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| | - Sadrettin Ekmen
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Neonatal Intensive Care Unit, Karabuk University, Karabuk, Turkey
| | - Eylem Sevınc
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Pediatric Gastroenterology Hepatology & Nutrition, Karabuk University, Karabuk, Turkey
| | - Erkan Dogan
- Department of Pediatrics, Faculty of Medicine Training and Research Hospital, Karabuk University, Karabuk, Turkey
| |
Collapse
|
233
|
Li Y, Zhang X, Yi J, Chen Y, Liang J, Wang L, Ma J, Zhu R, Zhang X, Hu D, Jia Y, Yu X, Wang Y. Synergistic evolution: The dynamic adaptation of SARS-CoV-2 and human protective immunity in the real world. J Infect 2024; 89:106310. [PMID: 39393556 DOI: 10.1016/j.jinf.2024.106310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES SARS-CoV-2 is continually evolving with new variants to evade protective immunity and cause new infections. This study aimed to assess infection-acquired immunity and hybrid immunity against re-infection or severe COVID-19. METHODS During 2020-2023, we collected 890 serum samples from individuals infected with SARS-CoV-2 variants including wild type, D614G, Alpha, Delta, BA.1, BA.2, BA.2.76, BA.5.2, BF.7, XBB, and EG.5. The levels of serum neutralizing antibodies (NAbs) against 18 diverse SARS-CoV-2 variants were determined using a bead-based high-throughput broad neutralizing-antibody assay. RESULTS In the initial wave of the COVID-19 pandemic, >75% of the patients demonstrated robust NAb responses against the ancestral SARS-CoV-2, during a period when vaccines were not yet available. After the emergence of the Omicron variant, the seroprevalence of anti-Omicron NAbs among the patients increased rapidly. By April 2023, when XBB variant was predominant, approximately 80% of the patients demonstrated >50% neutralization against the highly immune-evasive XBB lineages. Three serotypes of SARS-CoV-2, namely non-Omicron, Omicron, and XBB serotypes, were identified, with the strong likelihood of further changes occurring as the virus mutating. Generally, NAbs elicited by a previous serotype could not typically effectively protect against another serotype that emerges later in the evolutionary stages. CONCLUSION Our results firstly demonstrated the synergistic evolution between host immunity and SARS-CoV-2 variants in the real world, which would be helpful to develop future vaccines and public health strategies.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiaohan Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jingkun Yi
- Department of Biomedical Informatics, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yuan Chen
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Jing Liang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Li Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jiayue Ma
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Renlong Zhu
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Xiaomei Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Di Hu
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Yan Jia
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Xiaobo Yu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
234
|
Smidt-Nielsen IG, Boel JB, Meiniche H, Petersen A, Salgård C, Damkjær Bartels M, Holzknecht BJ. The added value of perineal swabs when screening for asymptomatic methicillin-resistant Staphylococcus aureus colonization and risk factors for perineal carriage. Diagn Microbiol Infect Dis 2024; 110:116537. [PMID: 39298934 DOI: 10.1016/j.diagmicrobio.2024.116537] [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: 06/24/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
We examined the added value of perineal swabs in addition to nose and throat swabs when screening for Methicillin-resistant Staphylococcus aureus colonization, and risk factors for perineal carriage in 6,642 patients. In our mainly primary care setting, the added value was 9.3%. Patients <3 or ≥80 had the highest risk.
Collapse
Affiliation(s)
- Isabel Guida Smidt-Nielsen
- Department of Clinical Microbiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Denmark.
| | - Jonas Bredtoft Boel
- Department of Clinical Microbiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark; Copenhagen University Hospital - The Hospital Pharmacy, Copenhagen, Denmark
| | - Heidi Meiniche
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Denmark
| | - Andreas Petersen
- Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Christian Salgård
- Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Mette Damkjær Bartels
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Barbara Juliane Holzknecht
- Department of Clinical Microbiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
235
|
Le DHH, Kanokudom S, Nguyen HM, Yorsaeng R, Honsawek S, Vongpunsawad S, Poovorawan Y. Hepatitis C Virus-Core Antigen: Implications in Diagnostic, Treatment Monitoring and Clinical Outcomes. Viruses 2024; 16:1863. [PMID: 39772172 PMCID: PMC11680303 DOI: 10.3390/v16121863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
The hepatitis C virus (HCV) infection, a global health concern, can lead to chronic liver disease. The HCV core antigen (HCVcAg), a viral protein essential for replication, offers a cost-effective alternative to HCV RNA testing, particularly in resource-limited settings. This review explores the significance of HCVcAg, a key protein in the hepatitis C virus, examining its structure, function, and role in the viral life cycle. It also evaluates its clinical use in diagnosis and treatment monitoring, comparing its performance to the standard HCV RNA assay using data from PubMed and Google Scholar. HCVcAg assays show high pooled sensitivity (93.5%) and pooled specificity (99.2%) compared to HCV RNA assays, correlating closely (r = 0.87) with HCV RNA levels. Hence, HCVcAg testing offers a cost-effective way to diagnose active HCV infections and monitor treatment, especially in resource-limited settings, but its sensitivity can vary and standardization is needed. HCVcAg also predicts liver disease progression and assesses liver damage risk, aiding patient management. It helps to identify patients at risk for fibrosis or carcinoma, making it vital in hepatitis C care. HCVcAg testing can expand access to HCV care, simplify management, and contribute to global elimination strategies, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Duong Hoang Huy Le
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (D.H.H.L.); (S.K.); (R.Y.); (S.V.)
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand;
- Medical Biochemistry & Molecular Biology Department, Fundamental Sciences and Basic Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam;
| | - Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (D.H.H.L.); (S.K.); (R.Y.); (S.V.)
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand;
| | - Ha Minh Nguyen
- Medical Biochemistry & Molecular Biology Department, Fundamental Sciences and Basic Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam;
- Laboratory Department, Nguyen Tri Phuong Hospital, Ho Chi Minh City 700000, Vietnam
| | - Ritthideach Yorsaeng
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (D.H.H.L.); (S.K.); (R.Y.); (S.V.)
| | - Sittisak Honsawek
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand;
| | - Sompong Vongpunsawad
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (D.H.H.L.); (S.K.); (R.Y.); (S.V.)
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (D.H.H.L.); (S.K.); (R.Y.); (S.V.)
- The Royal Society of Thailand, Sanam Sueapa, Bangkok 10330, Thailand
| |
Collapse
|
236
|
González-Navarro I, Urrea V, Gálvez C, Garcia-Guerrero MDC, Morón-López S, Puertas MC, Grau E, Mothe B, Bailón L, Miranda C, García F, Leal L, Vandekerckhove L, Marconi VC, Sekaly RP, Clotet B, Martinez-Picado J, Salgado M, the NIH Reversing Immune Dysfunction for HIV-1 Eradication (RID-HIV) Collaboratory Group. Assessing advances in three decades of clinical antiretroviral therapy on the HIV-1 reservoir. J Clin Invest 2024; 135:e183952. [PMID: 39610346 PMCID: PMC11735095 DOI: 10.1172/jci183952] [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: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUNDAntiretroviral therapy (ART) has improved the clinical management of HIV-1 infection. However, little is known about how the latest ART recommendations affect the heterogeneity of the HIV-1 reservoir size.METHODSWe used a complete statistical approach to outline parameters underlying the diversity in HIV-1 reservoir size in a cohort of 892 people with HIV-1 (PWH) on suppressive ART for more than 3 years. Total HIV-1-DNA levels were measured in PBMCs using digital droplet PCR (ddPCR).RESULTSWe classified 179 (20%) participants as being low viral reservoir treated (LoViReT) (<50 HIV-1-DNA copies/106 PBMCs). Twenty variables were collected to explore their association with the LoViReT phenotype using machine learning approaches. LoViReT status was closely associated with higher nadir CD4, lower zenith pre-ART viral load, lower CD4 recovery, shorter time from diagnosis to undetectable viral load, and initiation of treatment with an integrase inhibitor-containing (InSTI-containing) regimen. Initiation of ART with any InSTI was also linked with a shorter time to undetectable viremia. Locally estimated scatterplot smoothing (LOESS) regression revealed a progressive reduction in the size of the HIV-1 reservoir in individuals who started ART after 2007. Similarly, a higher nadir CD4 and a shorter time to undetectable viremia were observed when treatment was initiated after that year.CONCLUSIONOur findings demonstrate that the progressive implementation of earlier, universal treatment at diagnosis and the use of InSTIs affected the size of the HIV-1 reservoir. Our work shows that effective management of infection is the first step toward reducing the reservoir and brings us closer to achieving a cure.FUNDINGNIH; Division of AIDS at the National Institute of Allergy and Infectious Diseases (NIAID), NIH; Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Irene González-Navarro
- IrsiCaixa, Badalona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | | | | | | | - Sara Morón-López
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | - Maria C. Puertas
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | | | - Beatriz Mothe
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Lucía Bailón
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Cristina Miranda
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
| | - Felipe García
- Infectious Diseases Department Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Lorna Leal
- Infectious Diseases Department Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Vincent C. Marconi
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- The Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Rafick P. Sekaly
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
- Pathology Advanced Translational Research Unit (PATRU), Department of Pathology and Laboratory Medicine and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bonaventura Clotet
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Department of Infectious Diseases and Fundació Lluita contra les Infeccions, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
- Chair in Infectious Diseases and Immunity, University of Vic – Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Maria Salgado
- IrsiCaixa, Badalona, Barcelona, Spain
- Biomedical Research Networking Center on Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona, Spain
| | | |
Collapse
|
237
|
Lemos MP, Astronomo RD, Huang Y, Narpala S, Prabhakaran M, Mann P, Paez CA, Lu Y, Mize GJ, Glantz H, Westerberg K, Colegrove H, Smythe KS, Lin M, Pierce RH, Hutter J, Frank I, Mascola JR, McDermott AB, Bekker LG, McElrath MJ. Enhanced and sustained biodistribution of HIV-1 neutralizing antibody VRC01LS in human genital and rectal mucosa. Nat Commun 2024; 15:10332. [PMID: 39609400 PMCID: PMC11604655 DOI: 10.1038/s41467-024-54580-9] [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: 01/17/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
To prevent sexually-acquired HIV-1 infection by immunoprophylaxis, effective concentrations of broadly neutralizing antibodies are likely needed at mucosal sites of exposure. Here, we examine the biodistribution of monoclonal antibody VRC01 and its extended half-life variant, VRC01LS, in colorectal and genitourinary tracts of healthy adults 1-52 weeks after intravenous infusion. At 1-2 weeks, VRC01LS levels are ~3-4 times higher than VRC01 in serum (p = 0.048), rectal (p = 0.067), vaginal (p = 0.003) and cervical tissues (p = 0.003); these differences increase over time. Both antibodies primarily localize within rectal lamina propria and cervicovaginal stroma, with limited and variable epithelial distribution. Although 8-28% of serum mAb levels reach mucosal tissues, <3% are in seminal and rectal secretions. Elimination half-lives in mucosal tissues are 20-28 days for VRC01 and 51-68 days for VRC01LS. Thus, VRC01LS infusion achieves higher, sustained concentrations in human mucosal tissues than VRC01, supporting the future investigation of potent, long-acting LS-modified antibodies to prevent HIV-1.
Collapse
Affiliation(s)
- Maria P Lemos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rena D Astronomo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Madhu Prabhakaran
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Philipp Mann
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Carmen A Paez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gregory J Mize
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hayley Glantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katharine Westerberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hunter Colegrove
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kimberly S Smythe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Minggang Lin
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Robert H Pierce
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julia Hutter
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ian Frank
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Linda-Gail Bekker
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
238
|
Assayag E, Abulafia A, Teren D, Gelman E, Givoni H, Zadok D. Efficacy of 1% Povidone-Iodine in the Treatment of Anterior Blepharitis-Randomized Single-Center Controlled Trial. J Clin Med 2024; 13:7227. [PMID: 39685686 DOI: 10.3390/jcm13237227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Anterior blepharitis (AB) is a chronic eyelid inflammation with no definitive cure. Objectives: To assess the safety and efficacy of a 1% povidone-iodine (PVI) ophthalmic solution lid scrub compared to formulated eyelid wipes in treating AB. Design: A prospective, randomized, controlled, observer-masked, paired-eye trial. Methods: Sixty-three AB patients were randomly assigned to a 30-day treatment in which one eye underwent a daily lid scrub with 1% PVI solution (1% PVI group), while the fellow eye was treated with formulated eyelid wipes (control group). Clinical outcomes, such as blepharitis signs, tear breakup time, and corneal staining, were evaluated at study enrollment and exit visits. Symptom assessments utilized the visual analog scale (VAS) per eye and the ocular surface disease index (OSDI) questionnaire. Results: Fifty-two patients (mean age 62.3 years, 53.8% females) completed the treatment, while seven patients were lost to follow-up, three were non-compliant, and one sustained an eye trauma. After 30 days, both the 1% PVI and control groups exhibited significant improvements in symptoms, blepharitis signs, and corneal staining (p < 0.05). The 1% PVI scrubs were equally effective as eyelid wipes in most outcomes (p = 0.480) and superior in alleviating eyelid erythema (p = 0.007). Only the 1% PVI group showed a positive correlation between OSDI and VAS score improvements (r (52) = 0.353, p = 0.01). No adverse events related to either treatment modality were reported. Conclusions: A 1% PVI solution is an effective, safe, and well-tolerated treatment option for AB and is superior to formulated eyelid wipes in several subjective and objective measures.
Collapse
Affiliation(s)
- Elishai Assayag
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - Adi Abulafia
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - David Teren
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - Evgeny Gelman
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - Hila Givoni
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - David Zadok
- Department of Ophthalmology, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| |
Collapse
|
239
|
Blick-Nitko SK, Ture SK, Schafer XL, Munger JC, Livada AC, Li C, Maurya P, Rondina MT, Morrell CN. Platelet Ido1 expression is induced during Plasmodium yoelii infection, altering plasma tryptophan metabolites. Blood Adv 2024; 8:5814-5825. [PMID: 39133890 PMCID: PMC11609358 DOI: 10.1182/bloodadvances.2024013175] [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: 03/15/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 11/11/2024] Open
Abstract
ABSTRACT Platelets are immune responsive in many diseases as noted by changes in platelet messenger RNA in conditions such as sepsis, atherosclerosis, COVID-19, and many other inflammatory and infectious etiologies. The malaria causing Plasmodium parasite is a persistent public health threat and significant evidence shows that platelets participate in host responses to infection. Using a mouse model of nonlethal/uncomplicated malaria, non-lethal Plasmodium yoelii strain XNL (PyNL)-infected but not control mouse platelets expressed Ido1, a rate limiting enzyme in tryptophan metabolism that increases kynurenine at the expense of serotonin. Interferon-γ (IFN-γ) is a potent inducer of Ido1 and mice treated with recombinant IFN-γ had increased platelet Ido1 and IDO1 activity. PyNL-infected mice treated with anti-IFN-γ antibody had similar platelet Ido1 and metabolic profiles to that of uninfected controls. PyNL-infected mice become thrombocytopenic by day 7 after infection and transfusion of platelets from IFN-γ-treated wild-type mice but not Ido1-/- mice increased the plasma kynurenine-to-tryptophan ratio, indicating that platelets are a source of postinfection IDO1 activity. We generated platelet-specific Ido1 knockout mice to assess the contribution of platelet Ido1 during PyNL infection. Platelet-specific Ido1-/- mice had increased death and evidence of lung thrombi, which were not present in infected wild-type mice. Platelet Ido1 may be a significant contributor to plasma kynurenine in IFN-γ-driven immune processes and the loss of platelets may limit total Ido1, leading to immune and vascular dysfunction.
Collapse
Affiliation(s)
- Sara K. Blick-Nitko
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sara K. Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Xenia L. Schafer
- Department of Biochemistry, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Joshua C. Munger
- Department of Biochemistry, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Alison C. Livada
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Chen Li
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Preeti Maurya
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
240
|
Dudek B, Brożyna M, Karoluk M, Frankiewicz M, Migdał P, Szustakiewicz K, Matys T, Wiater A, Junka A. In Vitro and In Vivo Translational Insights into the Intraoperative Use of Antiseptics and Lavage Solutions Against Microorganisms Causing Orthopedic Infections. Int J Mol Sci 2024; 25:12720. [PMID: 39684431 DOI: 10.3390/ijms252312720] [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: 11/05/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
The growing antibiotic resistance of microorganisms causing postoperative infections following orthopedic surgeries underscores the urgent need for localized antiseptic and lavage delivery systems to enhance infection control. This study evaluates the in vitro effectiveness of antiseptic and lavage solutions-including polyhexanide, povidone-iodine, low-concentrated hypochlorite, Ringer's solution, and saline-against Staphylococcus epidermidis, Staphylococcus aureus MRSA, Cutibacterium acnes, Corynebacterium amycolatum, Pseudomonas aeruginosa, and Candida albicans. Using microplate models (Minimum Inhibitory Concentration, Minimum Biofilm Eradication Concentration, and Biofilm-Oriented Antiseptic Test assays), flow-based models (Bioflux system), and surfaces relevant to orthopedic implants (e.g., stainless steel disks/screws, Co-Cr-Mo, Ti-Al-Nb orthopedic alloys, and ultra-high-molecular-weight polyethylene), as well as a bio-nano-cellulose scaffold representing tissue, we assessed the solutions' activity. The cytotoxicity of the solutions was evaluated using osteoblast and keratinocyte cell lines, with additional in vivo insights gained through the Galleria mellonella larval model. The results show that polyhexanide-based solutions outperformed povidone-iodine in biofilm eradication in most tests applied, particularly on complex surfaces, whereas iodine demonstrated higher cytotoxicity in applied in vitro and in vivo tests. Low-concentration hypochlorite solutions exhibited minimal antibiofilm activity but also showed no cytotoxicity in cell line and G. mellonella larval models. These findings highlight the importance of careful antiseptic selection and rinsing protocols to balance infection control efficacy with tissue compatibility in orthopedic applications.
Collapse
Affiliation(s)
- Bartłomiej Dudek
- "P.U.M.A.", Platform for Unique Model Application, Department of Pharmacy, Wroclaw Medical University, Borowska 211, 50-534 Wroclaw, Poland
| | - Malwina Brożyna
- "P.U.M.A.", Platform for Unique Model Application, Department of Pharmacy, Wroclaw Medical University, Borowska 211, 50-534 Wroclaw, Poland
| | - Michał Karoluk
- Faculty of Mechanical Engineering, Department of Laser Technologies, Automation and Production Organization, Wrocław University of Science and Technology, Ignacego Łukasiewicza 5, 50-371 Wroclaw, Poland
| | - Mariusz Frankiewicz
- Faculty of Mechanical Engineering, Department of Laser Technologies, Automation and Production Organization, Wrocław University of Science and Technology, Ignacego Łukasiewicza 5, 50-371 Wroclaw, Poland
| | - Paweł Migdał
- Department of Bees Breeding, Institute of Animal Husbandry and Breeding, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Chełmońskiego 38C, 51-630 Wroclaw, Poland
| | - Konrad Szustakiewicz
- Department of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Tomasz Matys
- The Department and Clinic of Angiology and Internal Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Adrian Wiater
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Adam Junka
- "P.U.M.A.", Platform for Unique Model Application, Department of Pharmacy, Wroclaw Medical University, Borowska 211, 50-534 Wroclaw, Poland
| |
Collapse
|
241
|
Duchez AC, Arthaud CA, Eyraud MA, Prier A, Heestermans M, Hamzeh-Cognasse H, Cognasse F. Identification of new bioactive molecules in platelet preparation, storage, and transfusion reactions for improved transfusion management. Sci Rep 2024; 14:29381. [PMID: 39592728 PMCID: PMC11599570 DOI: 10.1038/s41598-024-80632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
Platelet concentrates (PCs) intended for transfusion contain bioactive molecules that can be considered Biological Response Modifiers (BRMs), mainly originating from plasma regardless of the preparation process. During storage, NGAL and GDF-15 levels increase in single donor apheresis platelet concentrates (SDA-PC), whereas in buffy coat platelet concentrates (BC-PC), the levels of MIP1α, MCP-3, and HSAA increase, and GDF-15 levels decrease. These molecules, primarily released by leukocytes, may contribute to adverse reactions (ARs) following a PC transfusion. Notably, in SDA-PC or BC-PC transfusions that result in ARs, the levels of NGAL, HSAA, and GDF-15 are significantly elevated, while the levels of MDC and CX3CL1 are significantly reduced compared to transfusions without ARs. These biomarkers could potentially serve as predictors for PCs-induced ARs.
Collapse
Affiliation(s)
- Anne-Claire Duchez
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France.
| | - Charles-Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| | - Marie-Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| | - Amélie Prier
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| | - Marco Heestermans
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| | - Hind Hamzeh-Cognasse
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
- INSERM, Université Jean Monnet, Mines Saint-Étienne, U 1059 SAINBIOSE, F- 42023, Saint-Etienne, France
| |
Collapse
|
242
|
Riccò M, Abu-Raya B, Icardi G, Spoulou V, Greenberg D, Pecurariu OF, Hung IFN, Osterhaus A, Sambri V, Esposito S. Respiratory Syncytial Virus: A WAidid Consensus Document on New Preventive Options. Vaccines (Basel) 2024; 12:1317. [PMID: 39771979 PMCID: PMC11679680 DOI: 10.3390/vaccines12121317] [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: 10/13/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Respiratory syncytial virus (RSV) is a leading cause of respiratory infections, particularly affecting young infants, older adults, and individuals with comorbidities. Methods: This document, developed as a consensus by an international group of experts affiliated with the World Association of Infectious Diseases and Immunological Disorders (WAidid), focuses on recent advancements in RSV prevention, highlighting the introduction of monoclonal antibodies (mAbs) and vaccines. Results: Historically, RSV treatment options were limited to supportive care and the monoclonal antibody palivizumab, which required multiple doses. Recent innovations have led to the development of long-acting mAbs, such as nirsevimab, which provide season-long protection with a single dose. Nirsevimab has shown high efficacy in preventing severe RSV-related lower respiratory tract infections (LRTIs) in infants, reducing hospitalizations and ICU admissions. Additionally, new vaccines, such as RSVpreF and RSVpreF3, target older adults and have demonstrated significant efficacy in preventing LRTIs in clinical trials. Maternal vaccination strategies also show promise in providing passive immunity to newborns, protecting them during the most vulnerable early months of life. This document further discusses the global burden of RSV, its economic impact, and the challenges of implementing these preventative strategies in different healthcare settings. Conclusions: The evidence supports the integration of both passive (mAbs) and active (vaccines) immunization approaches as effective tools to mitigate the public health impact of RSV. The combined use of these interventions could substantially reduce RSV-related morbidity and mortality across various age groups and populations, emphasizing the importance of widespread immunization efforts.
Collapse
Affiliation(s)
- Matteo Riccò
- Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy;
| | - Bahaa Abu-Raya
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority, Halifax, NS B3K 6R8, Canada;
- Departments of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Giancarlo Icardi
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece;
| | - David Greenberg
- Pediatric Infectious Diseases Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva 8410501, Israel;
| | - Oana Falup Pecurariu
- Children’s Clinical Hospital Brasov, 500063 Brasov, Romania;
- Faculty of Medicine Brasov, Transilvania University, 500019 Brasov, Romania
| | - Ivan Fan-Ngai Hung
- Division of Infectious Diseases, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR 999077, China;
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Vittorio Sambri
- Unit of Microbiology, The Greater Romagna Area Hub Laboratory, 47522 Cesena, Italy;
- Department Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
243
|
Tokarz-Deptuła B, Baraniecki Ł, Palma J, Stosik M, Deptuła W. Characterization of Platelet Receptors and Their Involvement in Immune Activation of These Cells. Int J Mol Sci 2024; 25:12611. [PMID: 39684330 DOI: 10.3390/ijms252312611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The article characterises platelets, pointing out the role and contribution of their numerous receptors determining their specific and broad immune activity. Three types of platelet receptors are described, that is, extracellular and intracellular receptors-TLR (toll-like receptors), NLR (NOD-like receptor), and RLR (RIG-I-like receptor); extracellular receptors-selectins and integrins; and their other extracellular receptors-CLR (C-type lectin receptor), CD (cluster of differentiation), TNF (tumour necrosis factor), among others. Outlining the contribution of these numerous platelet receptors to the intravascular immunity, it has been shown that they are formed by their fusion with pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and lifestyle-associated molecular patterns (LAMPs). They are initiating and effector components of signal transduction of these cells, and their expression and quantity determine the specific and broad functions of platelets towards influencing vascular endothelial cells, but mainly PRRs (pattern recognition receptors) of blood immune cells. These facts make platelets the fundamental elements that shape not only intravascular homeostasis, as previously indicated, but they become the determinants of immunity in blood vessels. Describing the reactions of the characterised three groups of platelet receptors with PAMP, DAMP and LAMP molecules, the pathways and participation of platelets in the formation and construction of intravascular immune status, in physiological states, but mainly in pathological states, including bacterial and viral infections, are presented, making these cells essential elements in the health and disease of mammals, including humans.
Collapse
Affiliation(s)
| | - Łukasz Baraniecki
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
- Doctoral School, University of Szczecin, 70-384 Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, 65-516 Zielona Góra, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
| |
Collapse
|
244
|
Baran A, Atılgan Lülecioğlu A, Gao L, Yazıcı YY, Demirel F, Metin A, Casanova JL, Puel A, Voyer TL, Beyaz Ş, Belkaya S. A Novel Heterozygous NFKB2 Variant in a Multiplex Family with Common Variable Immune Deficiency and Autoantibodies Against Type I IFNs. J Clin Immunol 2024; 45:48. [PMID: 39579251 PMCID: PMC11821294 DOI: 10.1007/s10875-024-01843-1] [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: 06/16/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
We studied a family with three male individuals across two generations affected by common variable immune deficiency (CVID). We identified a novel missense heterozygous variant (c.2602T>A:p.Y868N) of NFKB2 in all patients and not in healthy relatives. Functional studies of the mutant allele in an overexpression system and of the patients' cells confirmed the deleteriousness of the NFKB2 variant and genotype, respectively, on the activation of the non-canonical NF-κB signaling pathway. Impaired processing of p100 into p52 underlies p100 accumulation, which results in gain-of-function (GOF) of IκBδ inhibitory activity and loss-of-function (LOF) of p52 transcriptional activity. The three patients' plasma contained autoantibodies that neutralized IFN-α2 and/or IFN-ω, accounting for the severe or recurrent viral diseases of the patients, including influenza pneumonia in one sibling, and severe COVID-19 and recurrent herpes labialis in another. Our results confirm that NFKB2 alleles that are IκBδ GOF and p52 LOF can underlie CVID and drive the production of autoantibodies neutralizing type I IFNs, thereby predisposing to severe viral diseases.
Collapse
Affiliation(s)
- Alperen Baran
- Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara, Turkey
| | - Aysima Atılgan Lülecioğlu
- Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara, Turkey
| | - Liwei Gao
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Paris, France
| | - Yılmaz Yücehan Yazıcı
- Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara, Turkey
| | - Fevzi Demirel
- Department of Immunology and Allergy, Gülhane Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Ayşe Metin
- Division of Pediatric Immunology and Allergy Diseases, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Şengül Beyaz
- Division of Immunology and Allergy Diseases, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Serkan Belkaya
- Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara, Turkey.
- The National Nanotechnology Research Center (UNAM), İhsan Doğramacı Bilkent University, Ankara, Turkey.
| |
Collapse
|
245
|
Hamdan O, Amarin JZ, Potter M, Haddadin Z, Yanis A, Shawareb Y, Khuri-Bulos N, Haddadin R, Halasa NB, Spieker AJ. Seasonal influenza vaccination: Attitudes and practices of healthcare providers in Jordan. PLoS One 2024; 19:e0314224. [PMID: 39570858 PMCID: PMC11581314 DOI: 10.1371/journal.pone.0314224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Influenza is associated with significant global morbidity and mortality, with vaccination being the primary preventive strategy. Despite recommendations, influenza vaccine uptake among healthcare providers (HCPs) remains suboptimal, especially in the Eastern Mediterranean. We aimed to assess the attitudes and practices of HCPs in Jordan regarding seasonal influenza vaccination and assess sources of variation thereof. METHODS We conducted a cross-sectional survey study among a sample of HCPs practicing in Jordan (12/29/2020-04/26/2021). Participants completed a questionnaire assessing demographics, influenza vaccination history, attitudes, and practices. We used logistic regression to evaluate factors related to vaccine receipt and reasons for non-vaccination. We used proportional odds models to evaluate factors related to HCP recommendations and to compare opinions on influenza vaccination between ever- and never-vaccinated HCPs. RESULTS Of 305 survey initiates, 206 HCPs (67.5%) comprised the analytic sample. The median age was 35 years; 61.2% were women, and 43.7% were pharmacists. Over half (52.9%) never received an influenza vaccine; however, older age and self-identifying as a physician were associated with higher odds of having ever received the influenza vaccine. The main reasons for non-vaccination were related to the misassessment of risks and benefits. Prior receipt of influenza vaccination was strongly associated with odds of recommending vaccination (or = 10.5; 95% CI = [5.38-20.3]; p<0.001). The COVID-19 pandemic reportedly enhanced influenza vaccine acceptance among 48.5% of HCPs surveyed. CONCLUSIONS Low influenza vaccine uptake among healthcare providers in Jordan is related to misassessment of risks and benefits. Enhancing attitudes and confidence through tailored education is crucial to overcoming hesitancy and promoting sustained improvements in vaccination attitudes and practices among HCPs in Jordan.
Collapse
Affiliation(s)
- Olla Hamdan
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Justin Z. Amarin
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Epidemiology Doctoral Program, School of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Molly Potter
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Zaid Haddadin
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Ahmad Yanis
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Yanal Shawareb
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Najwa Khuri-Bulos
- Department of Pediatrics, School of Medicine, The University of Jordan, Amman, Jordan
| | - Randa Haddadin
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Natasha B. Halasa
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Andrew J. Spieker
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
246
|
Luo F, Xu C, Zhang C, Tan A, Lu D, Luo P, Cheng P, Zhang W, Bai L, Yu C, Sun S, Zeng H, Zou Q. mRNA-based platform for preventing and treating Staphylococcus aureus by targeted staphylococcal enterotoxin B. Front Immunol 2024; 15:1490044. [PMID: 39640268 PMCID: PMC11617584 DOI: 10.3389/fimmu.2024.1490044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus (S. aureus) possesses numerous virulence factors, with the increasing prevalence of drug-resistant strains heightening the threat posed by this pathogen. Staphylococcal enterotoxin B (SEB), a highly conserved toxin secreted by S. aureus, is also recognized as a potential bioweapon with super-antigenic activity. SEB represents a promising target in efforts to combat infections caused by S. aureus. We developed mRNA-based vaccine and antibody targeting SEB for both prophylactic and therapeutic purposes in varying S. aureus infection conditions. The mSEB mRNA vaccine (10 μg per mouse) induces more robust and persistent immune responses, including higher antibody titers and specific cellular immune responses, compared to immunization with 30 μg of mSEB protein adjuvanted with aluminum phosphate. Additionally, the anti-SEB mRNA antibody maintains secretion of anti-SEB monoclonal antibody (mAb) with a dosage that is 10 times lower than purified protein administration. The mRNA-based antibody exhibits superior pharmacokinetic profiles compared to its protein counterparts, efficiently neutralizing SEB and clearing S. aureus from circulation. Both the mRNA vaccine and mRNA antibody demonstrate preventive and therapeutic effects by eliciting specific immune responses and generating high-affinity antibodies in mice. We have laid the groundwork for the development and evaluation of mRNA-based vaccines and antibodies targeting SEB produced by S. aureus. Our studies demonstrate that these approaches are more effective than traditional protein-based vaccines and antibodies in terms of inducing immune responses, pharmacokinetics, and their prophylactic or therapeutic efficacy against S. aureus infections.
Collapse
Affiliation(s)
- Fumei Luo
- School of Pharmacy, University of South China, Hunan, China
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chengwen Zhang
- Medical Research Institute, Southwest University, Chongqing, China
| | - Aomo Tan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Lijuan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Cuiyun Yu
- School of Pharmacy, University of South China, Hunan, China
| | - Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| |
Collapse
|
247
|
Yamada S, Harada S, Fujii H, Kinoshita H, Nguyen PHA, Shibamura M, Yoshikawa T, Kawahara M, Ebihara H, Saijo M, Fukushi S. A Mutation in the Herpes Simplex Virus Type 1 (HSV-1) UL29 Gene is Associated with Anti-Herpesvirus Drugs' Susceptibility. Viruses 2024; 16:1813. [PMID: 39772124 PMCID: PMC11680290 DOI: 10.3390/v16121813] [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: 08/29/2024] [Revised: 10/18/2024] [Accepted: 11/20/2024] [Indexed: 01/25/2025] Open
Abstract
Herpes simplex virus type 1 (HSV-1) acyclovir (ACV) resistance is acquired by mutations in the viral thymidine kinase (TK) or DNA polymerase (DNApol) genes. We previously obtained an ACV-resistant clone (HSV-1_VZV_TK_clone α) by sequential passages of HSV-1_VZV-TK, a recombinant virus which lacked its endogenous TK activity and instead expressed the varicella-zoster virus (VZV) TK ectopically. HSV-1_VZV_TK_clone α had been generated using an HSV-1_BAC in the presence of increasing concentrations of ACV. The ACV-resistant clone bore normal TK and DNApol genes. Here, we deployed next-generation full-genome sequencing of HSV-1_VZV_TK_clone α and identified a single nucleotide substitution, resulting in a P597L missense mutation in the UL29 gene product, the ICP8 protein. Recombinant HSV-1 encoding a P597L ICP8 protein was generated, and its properties and ability to confer drug resistance were analyzed. No difference in virus growth and UL29 expression was observed between the mutant recombinant, the wild type, and a revertant mutant viral strain, and susceptibility tests of these strains to ACV and other drugs using Vero, HEL, and ARPE19 cells identified that the recombinant UL29 mutant virus was resistant only to ACV. These results indicate that ICP8 may be involved in the anti-herpesvirus drugs' mechanism of action on HSV-1.
Collapse
Grants
- 21591402, 24591591, 15K09675, 15K19594, 18K07894, 20K06404, and 21K05967 Ministry of Education, Culture, Sports, Science, and Technology of Japan
- H21-Shinko-Ippan-009 Ministry of Health, Labor and Welfare Science Research Grant
Collapse
Affiliation(s)
- Souichi Yamada
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Shizuko Harada
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Hikaru Fujii
- The Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Ehime, Japan;
| | - Hitomi Kinoshita
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Phu Hoang Anh Nguyen
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Miho Shibamura
- Center for Surveillance, Immunization and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo 162-8640, Japan;
| | - Tomoki Yoshikawa
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Madoka Kawahara
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Hideki Ebihara
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
- Health and Welfare Bureau, Sapporo City 060-8611, Hokkaido, Japan
| | - Shuetsu Fukushi
- Department of Virology 1, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (S.H.); (H.K.); (P.H.A.N.); (T.Y.); (M.K.); (H.E.); (M.S.); (S.F.)
| |
Collapse
|
248
|
Shen N, Wu R, Lu T, Jiang Y, Ning T, Liu S, Liu X, Zhu S, Qiao J. Characterization of oral microbiota of children with hand, foot, and mouth disease caused by enterovirus A 71. BMC Infect Dis 2024; 24:1331. [PMID: 39574007 PMCID: PMC11583527 DOI: 10.1186/s12879-024-10233-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The association between alterations in the oral microbiome and hand, foot, and mouth disease (HFMD) has been observed in previous studies. Our study, therefore, aimed to identify the structural changes in the oral microbiota and biomarkers in children with HFMD caused by enterovirus A 71 (EV-A71). METHODS Children diagnosed with EV-A71 HFMD and healthy children recruited from April 2021 to September 2023 were included in the present study, and were categorized into EV-A71 and control groups, respectively. Oral swabs were collected and microbiota information was obtained using 16 S rRNA gene sequencing technology. Alpha-diversity and partial least squares discriminant analyses were conducted to compare microbial diversity, richness, and similarity between the two groups. Linear discriminant analysis effect size was employed to identify microbial taxa with significant differences, and determined the key genera among them. RESULTS The study included a total of 80 children, with 50 assigned to the EV-A71 group and 30 to the control group. No significant differences were found between the two groups in terms of age (2.2 ± 1.2 vs. 2.7 ± 1.2 years; age range: 1-5 years; P = 0.114) or sex (56% vs. 60% boys, P = 0.726). The oral microbiota structure in the EV-A71 group differed from that in the control group. The EV-A71 group showed significant reductions in both the Shannon index (P = 0.037) and the abundance-based coverage estimator (ACE) index (P < 0.001). The key genus changes were marked by a significant decrease in the abundance of Capnocytophaga (P = 0.002) and Leptotrichia (P = 0.033) in the EV-A71 group. CONCLUSION In children with EV-A71 HFMD, the oral microbiota showed changes in composition, with a significant reduction in diversity and richness. The changes in key genera were a marked decrease in the abundance of Capnocytophaga and Leptotrichia.
Collapse
Affiliation(s)
- Nan Shen
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Rang Wu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Tiantian Lu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Yazhou Jiang
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Tao Ning
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Song Liu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Xiang Liu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China
| | - Suyue Zhu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China.
| | - Jibing Qiao
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu, 223800, People's Republic of China.
| |
Collapse
|
249
|
Liau SK, Hung CC, Chen CY, Liu YC, Lu YA, Lin YJ, Chen YC, Tian YC, Tseng FG, Hsu HH. A Quick Sequential Organ Failure Assessment (qSOFA) Score Greater than 1 and Shortened Ampicillin Use Predict Death and One-Year Mortality in Hospitalized Patients with Non-Perinatal Invasive Listeriosis: A Retrospective Analysis of 118 Consecutive Cases. Microorganisms 2024; 12:2365. [PMID: 39597753 PMCID: PMC11596919 DOI: 10.3390/microorganisms12112365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Listeria monocytogenes causes listeriosis, a serious foodborne illness with a high mortality rate, especially in vulnerable populations. It accounts for 19% of foodborne deaths, with invasive cases having a mortality rate of up to 44%, leading to conditions like meningitis, bacteremia, and meningoencephalitis. However, the prognostic factors remain unclear. This study examines the hospital outcomes of invasive listeriosis and identifies risk factors for in-hospital and one-year mortality. We analyzed the electronic medical records of 118 hospitalized patients with non-perinatal, culture-proven invasive listeriosis collected over a 21-year period. The in-hospital mortality rate was 36.4%, with only 33.1% surviving one year and 22.0% surviving two years. The key findings indicate that a quick Sequential Organ Failure Assessment (qSOFA) score of ≥2 (OR 106.59, p < 0.001), respiratory failure (OR 7.58, p = 0.031), and shorter ampicillin duration (OR 0.53, p = 0.012) independently predicted poorer in-hospital outcomes. Additionally, a qSOFA score of ≥2 (OR 8.46, p < 0.001) and shorter ampicillin duration (OR 0.65, p < 0.001) were linked to higher one-year mortality. This study is the first to identify a qSOFA score of ≥2 as a significant marker for high-risk invasive listeriosis patients, with poorer outcomes linked to a qSOFA score of ≥2, respiratory failure, and shorter ampicillin use.
Collapse
Affiliation(s)
- Shuh-Kuan Liau
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Cheng-Chieh Hung
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Chao-Yu Chen
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Yi-Chun Liu
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Yueh-An Lu
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Yu-Jr Lin
- Research Services Center for Health Information, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Yung-Chang Chen
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Ya-Chung Tian
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| | - Fan-Gang Tseng
- Department of Engineering and System Science, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Institute of Nano Engineering and Microsystems, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsiang-Hao Hsu
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Branch, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (S.-K.L.); (C.-Y.C.); (Y.-C.L.); (Y.-A.L.); (Y.-C.C.); (Y.-C.T.)
| |
Collapse
|
250
|
Xiao L, Ma J, Chen R, Chen J, Wang Q, Tang N, Zhao X, Zhang H, Jiao C. The Impact of Cytomegalovirus Infection on Ulcerative Colitis Relapse: A Multicenter Retrospective Cohort Study. J Inflamm Res 2024; 17:9059-9070. [PMID: 39583855 PMCID: PMC11585274 DOI: 10.2147/jir.s479663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
Purpose Cytomegalovirus (CMV) infection exacerbates intestinal inflammation in ulcerative colitis (UC) patients, yet the effect of CMV infection on UC relapse has not been fully elucidated. This study aimed to investigate the impact of CMV infection on UC relapse and identify associated risk factors. Patients and Methods This multicenter retrospective cohort study included UC patients who visited research centers from January 2016 to December 2020. Univariate and multivariate Cox regression analyses were conducted to explore risk factors for UC relapse. Propensity score matching was used to balance the differences in the clinical characteristics between the groups. Results A total of 298 UC patients participated in this study, including 19 with CMV colitis, 37 with CMV viremia, and 242 CMV-negative patients. The 2-year cumulative recurrence rate was higher in patients with CMV colitis than that in CMV-negative patients (84.21% vs 51.65%, p = 0.01). Univariate and multivariate Cox regression analyses confirmed that fecal calprotectin ≥ 250 µg/g, Montreal classification E3, CMV colitis, duration > 48 months, and serum albumin < 30 g/L were independent risk factors for UC relapse at 2 years, whereas the use of biologics for induction of remission was identified as an independent protective factor. Conclusion Our study suggests that the risk of relapse increases among UC patients with CMV colitis over two years. Risk factors for UC relapse at 2 years include fecal calprotectin ≥ 250 μg/g, Montreal classification E3, CMV colitis, UC duration > 48 months, and albumin < 30 g/L, whereas the use of biologics during induction is a protective factor.
Collapse
Affiliation(s)
- Linmei Xiao
- Department of Liver Disease, Wuxi No.5 People’s Hospital Affiliated to Jiangnan University, Wuxi, People’s Republic of China
| | - Jingjing Ma
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Ruidong Chen
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Jie Chen
- Northern Jiangsu People’s Hospital/Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, Jiangsu Province, People’s Republic of China
| | - Qiang Wang
- Jiangsu Shengze Hospital, Suzhou, Jiangsu Province, People’s Republic of China
| | - Nana Tang
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Xiaojing Zhao
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Hongjie Zhang
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Chunhua Jiao
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| |
Collapse
|