101
|
Thieulent CJ, Balasuriya UBR, Tseng A, Crossland NA, Stephens JM, Dittmar W, Staszkiewicz J, Richt JA, Carossino M. Diabetes exacerbates SARS-CoV-2 replication through ineffective pulmonary interferon responses, delayed cell-mediated immunity, and disruption of leptin signaling. Front Cell Infect Microbiol 2025; 15:1513687. [PMID: 40125513 PMCID: PMC11925909 DOI: 10.3389/fcimb.2025.1513687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/25/2025] Open
Abstract
Comorbidities, including obesity and type 2 diabetes mellitus (T2DM), are associated with increased disease severity and mortality following SARS-CoV-2 infection. Here, we investigated virus-host interactions under the effects of these comorbidities in diet-induced obesity (DIO) and leptin receptor-deficient (T2DM) mice following infection with SARS-CoV-2. DIO mice, as well as their lean counterparts, showed limited susceptibility to SARS-CoV-2 infection. In contrast, T2DM mice showed exacerbated pulmonary SARS-CoV-2 replication and delayed viral clearance associated with down-regulation of innate and adaptative immune gene signatures, ineffective type I interferon response, and delayed SARS-CoV-2-specific cell-mediated immune responses. While T2DM mice showed higher and prolonged SARS-CoV-2-specific immunoglobulin isotype responses compared to their lean counterparts, neutralizing antibody levels were equivalent. By silencing the leptin receptor in vitro using a human alveolar epithelial cell line, we observed an increase in SARS-CoV-2 replication and type I interferons. Altogether, our data provides for the first time evidence that disruption of leptin receptor signaling leading to obesity and T2DM induces altered type I interferon and cell-mediated responses against SARS-CoV-2, mediating increased viral replication and delayed clearance. These data shed light on the alteration of the innate immune pathway in the lung using in-depth transcriptomic analysis and on adaptive immune responses to SARS-CoV-2 under T2DM conditions. Finally, this study provides further insight into this risk factor aggravating SARS-CoV-2 infection and understanding the underlying cellular mechanisms that could help identify potential intervention points for this at-risk population.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/physiology
- SARS-CoV-2/immunology
- Mice
- COVID-19/immunology
- COVID-19/virology
- Virus Replication
- Receptors, Leptin/genetics
- Receptors, Leptin/metabolism
- Obesity/immunology
- Obesity/complications
- Signal Transduction
- Humans
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/complications
- Leptin/metabolism
- Interferon Type I/immunology
- Interferon Type I/metabolism
- Lung/immunology
- Lung/virology
- Immunity, Cellular
- Mice, Inbred C57BL
- Immunity, Innate
- Male
- Disease Models, Animal
- Antibodies, Neutralizing/blood
- Interferons
- Mice, Knockout
Collapse
Affiliation(s)
- Côme J. Thieulent
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Anna Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Wellesley Dittmar
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jaroslaw Staszkiewicz
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Juergen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
102
|
Kordi R, Chang AJ, Hicar MD. Serology supportive of recent coxsackievirus B infection is correlated with multisystem inflammatory syndrome in children (MIS-C). Microbiol Spectr 2025; 13:e0174124. [PMID: 39907434 PMCID: PMC11878073 DOI: 10.1128/spectrum.01741-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Rarely, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will lead to myocarditis associated with multisystem inflammatory syndrome in children (MIS-C). It remains unclear why MIS-C only targets specific children. To explore an association between coxsackievirus infections with MIS-C, we investigated the sero-epidemiology of CV in admitted pediatric patients in relation to the pandemic. This retrospective case-control study was performed by chart review of children (age ≤21 years) admitted to a tertiary care hospital with CV serological testing from January 2017 to August 2023. Clinical, laboratory, and imaging findings were used to classify patients as MIS-C and CV-unlikely or CV-possible for non-MIS-C patients. Out of 182 admissions (179 patients, median age, 6), CVB complement fixation (CF) assay on serotypes B1-B6 and CVA immunofluorescence assay IgG on serotypes A7, A9, A16, and A24 were positive in at least one serotype in 59.2% and 80.7% of cases, respectively. We observed a significant drop in CVB CF seropositivity during the peak of social distancing in 2020. The likelihood of elevated CVB CF titers was significantly higher in MIS-C than the CV-unlikely group (OR: 1.92, 95% CI: 1.02-3.63, P: 0.04) and showed a trend toward higher values in African Americans than Whites (OR: 1.57, 95% CI: 0.98-2.50, P: 0.057). The frequency of MIS-C was considerably higher in African Americans than Whites (18.1% versus 9%, P: 0.1). A higher likelihood of elevated CVB CF titers in patients with MIS-C compared with those unlikely to have acute CV infection along with a relatively higher frequency of MIS-C in African Americans warrants further investigation into the role of CVB infection in MIS-C development.IMPORTANCEThe emergence of multisystem inflammatory syndrome in children (MIS-C) during the SARS-CoV-2 pandemic raised major concerns in providers caring for children. This condition presents a hyper-inflammation state that can lead to severe complications, including myocarditis and cardiogenic shock. The pathogenesis of MIS-C has not been fully understood. Understanding the pathogenesis of this condition is not only important for developing effective treatments but also for applying preventive strategies. A two-hit hypothesis leading to MIS-C has been proposed. Coxsackievirus infections are prevalent during childhood and can also cause myocarditis, and coxsackievirus B specifically has been shown to cause persistent RNA presence in host cells, leading to continued inflammation. Herein, we show that elevated coxsackievirus B titers are associated with MIS-C cases, implying a role of successive infections with these viruses contributing to such a hyperinflammatory state. This study supports the need for larger investigations into this association.
Collapse
Affiliation(s)
- Ramesh Kordi
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, New York, USA
| | - Arthur J. Chang
- Division of Pediatric Infectious Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mark D. Hicar
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
103
|
Ma Y, Du Y, Yang J, Wang H, Lin X. Effect of Inactivated Vaccines Against SARS-CoV-2 on Immunogenicity Outcome. Disaster Med Public Health Prep 2025; 19:e50. [PMID: 40033891 DOI: 10.1017/dmp.2024.331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
OBJECTIVES The purpose of this study was to measure and examine the levels of IgG, IgM, and Spike antibody induced by inactivated vaccines, including CoronaVac and BBIBP-CorV. METHODS Two groups of healthy adults over 18 years old (50 participants per group), who had previously received 1 dose of either BBIBP-CorV or CoronaVac and receiving either a homologous booster of BBIBP-CorV or a heterologous booster of CoronaVac. Serum IgG, IgM, and Spike antibody levels against SARS-COV-2 were measured using magnetic particle chemiluminescence immunoassay and the ELISA method. RESULTS The results showed that both spike antibody and IgG/IgM antibodies elicited by a CoronaVac booster following 1 dose of BBIBP-CorV were significantly higher than those elicited by either a homologous BBIBP-CorV booster or a heterologous BBIBP-CorV booster. The Spike antibody against SARS-COV-2 induced by the heterologous CoronaVac booster reached 200.3, which is substantially greater than that induced by the homologous BBIBP-CorV booster (127.5 pg/mL). Conversely, the Spike antibody against SARS-COV-2 induced by the heterologous BBIBP-CorV booster reached 53.93 pg/mL, which is substantially greater than that induced by the homologous CoronaVac booster (9.60 pg/mL). CONCLUSIONS In summary, CoronaVac is immunogenic as a booster dose following 1 dose of BBIBP-CorV and is immunogenically superior to both the homologous booster and the heterologous BBIBP-CorV booster.
Collapse
Affiliation(s)
- Yuke Ma
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yukuan Du
- Department of Clinical Laboratory, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jingnan Yang
- Department of Clinical Laboratory, Huaihe Hospital of Henan University, Kaifeng, China
| | - Huichao Wang
- Department of Nephrology, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Xuhong Lin
- Department of Clinical Laboratory, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
104
|
Ratti M, Rescinito R, Gigante D, Lontano A, Panella M. Influenza Vaccination of Nurses and Other Health Care Workers in Different Occupational Settings: A Classic and AI Mixed Approach for Time-to-Event Data. NURSING REPORTS 2025; 15:87. [PMID: 40137660 PMCID: PMC11944323 DOI: 10.3390/nursrep15030087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Seasonal influenza currently remains a major public health concern for the community and, in particular, the health care worker (HCW). According to the World Health Organization, HCWs are among the high-risk categories for which vaccination is recommended, due to the derived absenteeism, productivity loss, and high probability of transmitting the disease to vulnerable individuals or patients. Therefore, an HCW vaccination policy should be adopted by every health care provider. There is growing evidence that a time effect of the vaccination event is probable, which may influence vaccine effectiveness. We designed and conducted an observational study to investigate the time to anti-influenza vaccination event of different categories of HCWs belonging to different occupational settings in a tertiary hospital during three seasons in order to retrieve some insight about HCW prioritization when designing vaccination campaigns. Materials and Methods: We retrospectively analyzed the results of two HCW anti-influenza vaccination campaigns (2022 and 2023) to assess any difference regarding job typology and unit typology (critical care, surgical, medical, service). We first fitted a classic Cox proportional hazard model and then an AI random forest model to assess variable importance. We used R, RStudio, and the survex package. Results: Overall, other HCWs reported a lower vaccination rate compared to nurses (HR 0.77; 95%CI 0.62-0.97), and service unit personnel appeared to more likely be vaccinated (HR 1.42; 95%CI 1.01-1.99) compared to those belonging to the critical care units. As expected, older workers tended to be vaccinated more frequently (HR 1.70 for the (46, 65] category compared to the younger one; 95%CI 1.39-2.09). The variable importance analysis showed consistent superiority of the ward typology and age category variables with respect to time. During the entire timeline, the ward typology appeared to be more important than the HCW typology. Conclusions: Our results suggest a prioritization policy based firstly on the unit typology followed by the job typology for HCW anti-influenza campaigns.
Collapse
Affiliation(s)
- Matteo Ratti
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale, 28100 Novara, Italy; (R.R.); (D.G.); (M.P.)
- Azienda Ospedaliero Universitaria Maggiore della Carità, 28100 Novara, Italy;
| | - Riccardo Rescinito
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale, 28100 Novara, Italy; (R.R.); (D.G.); (M.P.)
- Azienda Ospedaliero Universitaria Maggiore della Carità, 28100 Novara, Italy;
| | - Domenico Gigante
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale, 28100 Novara, Italy; (R.R.); (D.G.); (M.P.)
- Occupational Safety and Prevention Service (SPreSAL), ASL VC (Local Health Authority), 13100 Vercelli, Italy
| | - Alberto Lontano
- Azienda Ospedaliero Universitaria Maggiore della Carità, 28100 Novara, Italy;
- Section of Hygiene, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimiliano Panella
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale, 28100 Novara, Italy; (R.R.); (D.G.); (M.P.)
- Azienda Ospedaliero Universitaria SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| |
Collapse
|
105
|
Fritsch KJ, Krüger L, Handtke S, Kohler TP, Ozhiganova A, Jahn K, Wesche J, Greinacher A, Hammerschmidt S. Pneumococcal Neuraminidases Increase Platelet Killing by Pneumolysin. Thromb Haemost 2025; 125:243-254. [PMID: 39029905 DOI: 10.1055/a-2369-8680] [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] [Indexed: 07/21/2024]
Abstract
BACKGROUND Platelets prevent extravasation of capillary fluids into the pulmonary interstitial tissue by sealing gaps in inflamed endothelium. This reduces respiratory distress associated with pneumonia. Streptococcus pneumoniae is the leading cause of severe community-acquired pneumonia. Pneumococci produce pneumolysin (PLY), which forms pores in membranes of eukaryotic cells including platelets. Additionally, pneumococci express neuraminidases, which cleave sialic acid residues from eukaryotic glycoproteins. In this study, we investigated the effect of desialylation on PLY binding and pore formation on platelets. MATERIALS AND METHODS We incubated human platelets with purified neuraminidases and PLY, or nonencapsulated S. pneumoniae D39/TIGR4 and isogenic mutants deficient in PLY and/or NanA. We assessed platelet desialylation, PLY binding, and pore formation by flow cytometry. We also analyzed the inhibitory potential of therapeutic immunoglobulin G preparations (IVIG [intravenous immunoglobulin]). RESULTS Wild-type pneumococci cause desialylation of platelet glycoproteins by neuraminidases, which is reduced by 90 to 100% in NanA-deficient mutants. NanC, cleaving only α2,3-linked sialic acid, induced platelet desialylation. PLY binding to platelets then x2doubled (p = 0.0166) and pore formation tripled (p = 0.0373). A neuraminidase cleaving α2,3-, α2,6-, and α2,8-linked sialic acid like NanA was even more efficient. Addition of polyvalent IVIG (5 mg/mL) decreased platelet desialylation induced by NanC up to 90% (p = 0.263) and reduced pore formation >95% (p < 0.0001) when incubated with pneumococci. CONCLUSION Neuraminidases are key virulence factors of pneumococci and desialylate platelet glycoproteins, thereby unmasking PLY-binding sites. This enhances binding of PLY and pore formation showing that pneumococcal neuraminidases and PLY act in concert to kill platelets. However, human polyvalent immunoglobulin G preparations are promising agents for therapeutic intervention during severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Kristin J Fritsch
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Laura Krüger
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Rostock, Rostock, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Arina Ozhiganova
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Kristin Jahn
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Jan Wesche
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
106
|
de Oliveira Silva Pinto M, de Paula Pereira L, de Mendonça Angelo ALP, Xavier MAP, de Magalhães Vieira Machado A, Russo RC. Dissecting the COVID-19 Immune Response: Unraveling the Pathways of Innate Sensing and Response to SARS-CoV-2 Structural Proteins. J Mol Recognit 2025; 38:e70002. [PMID: 39905998 DOI: 10.1002/jmr.70002] [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: 11/08/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), the virus responsible for COVID-19, interacts with the host immune system through complex mechanisms that significantly influence disease outcomes, affecting both innate and adaptive immunity. These interactions are crucial in determining the disease's severity and the host's ability to clear the virus. Given the virus's substantial socioeconomic impact, high morbidity and mortality rates, and public health importance, understanding these mechanisms is essential. This article examines the diverse innate immune responses triggered by SARS-CoV-2's structural proteins, including the spike (S), membrane (M), envelope (E), and nucleocapsid (N) proteins, along with nonstructural proteins (NSPs) and open reading frames. These proteins play pivotal roles in immune modulation, facilitating viral replication, evading immune detection, and contributing to severe inflammatory responses such as cytokine storms and acute respiratory distress syndrome (ARDS). The virus employs strategies like suppressing type I interferon production and disrupting key antiviral pathways, including MAVS, OAS-RNase-L, and PKR. This study also explores the immune pathways that govern the activation and suppression of immune responses throughout COVID-19. By analyzing immune sensing receptors and the responses initiated upon recognizing SARS-CoV-2 structural proteins, this review elucidates the complex pathways associated with the innate immune response in COVID-19. Understanding these mechanisms offers valuable insights for therapeutic interventions and informs public health strategies, contributing to a deeper understanding of COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Matheus de Oliveira Silva Pinto
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo de Paula Pereira
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
107
|
Lukeman H, Al-Wassiti H, Fabb SA, Lim L, Wang T, Britton WJ, Steain M, Pouton CW, Triccas JA, Counoupas C. An LNP-mRNA vaccine modulates innate cell trafficking and promotes polyfunctional Th1 CD4 + T cell responses to enhance BCG-induced protective immunity against Mycobacterium tuberculosis. EBioMedicine 2025; 113:105599. [PMID: 39955975 PMCID: PMC11871481 DOI: 10.1016/j.ebiom.2025.105599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Mycobacterium tuberculosis remains the largest infectious cause of mortality worldwide, even with over a century of widespread administration of the only licenced tuberculosis (TB) vaccine, Bacillus Calmette-Guérin (BCG). mRNA technology remains an underexplored approach for combating chronic bacterial infections such as TB. METHODS We have developed a lipid nanoparticle (LNP)-mRNA vaccine, termed mRNACV2, encoding for the M. tuberculosis CysVac2 fusion protein, which we have previously formulated as an adjuvanted subunit vaccine. This LNP-mRNA vaccine was administered intramuscularly to female C57BL/6 mice as a standalone vaccine or as booster to BCG to assess immunogenicity and efficacy of the construct. FINDINGS Vaccination with mRNACV2 induced high frequencies of polyfunctional, antigen-specific Th1 CD4+ T cells in the blood and lungs, which was associated with the rapid recruitment of both innate and adaptive immune cells to lymph nodes draining the site of immunisation. mRNACV2 vaccination also provided significant pulmonary protection in M. tuberculosis-infected mice, reducing bacterial load and inflammatory infiltration in the lungs. Importantly, mRNACV2 enhanced immune responses and long-term protection when used to boost BCG-primed mice. INTERPRETATION These findings of a protective LNP-mRNA vaccine for TB highlight the potential of the LNP-mRNA platform for TB control and support further research to facilitate translation to humans. FUNDING This work was supported by the NHMRC Centre of Research Excellence in Tuberculosis Control to JAT and WJB (APP1153493), and MRFF mRNA Clinical Trial Enabling Infrastructure grant to CWP and HAW (MRFCTI000006).
Collapse
Affiliation(s)
- Hannah Lukeman
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Hareth Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Trixie Wang
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Warwick J Britton
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Megan Steain
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - James A Triccas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Claudio Counoupas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
108
|
Fang X, Mo C, Zheng L, Gao F, Xue F, Zheng X. Transfusion-Related Acute Lung Injury: from Mechanistic Insights to Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413364. [PMID: 39836498 PMCID: PMC11923913 DOI: 10.1002/advs.202413364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/08/2024] [Indexed: 01/23/2025]
Abstract
Transfusion-related acute lung injury (TRALI) is a potentially lethal complication of blood transfusions, characterized by the rapid onset of pulmonary edema and hypoxemia within six hours post-transfusion. As one of the primary causes of transfusion-related mortality, TRALI carries a significant mortality rate of 6-12%. However, effective treatment strategies for TRALI are currently lacking, underscoring the urgent need for a comprehensive and in-depth understanding of its pathogenesis. This comprehensive review provides an updated and detailed analysis of the current landscape of TRALI, including its clinical presentation, pathogenetic hypotheses, animal models, cellular mechanisms, signaling pathways, and potential therapeutic targets. By highlighting the critical roles of these pathways and therapies, this review offers valuable insights to inform the development of preventative and therapeutic strategies and to guide future research efforts aimed at addressing this life-threatening condition.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology/Critical Care MedicineFuzhou University Affiliated Provincial HospitalSchool of MedicineFuzhou UniversityShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial HospitalFuzhouFujian350001China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOEState Key Laboratory of BiotherapyWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Ling Zheng
- Department of Anesthesiology/Critical Care MedicineFuzhou University Affiliated Provincial HospitalSchool of MedicineFuzhou UniversityShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial HospitalFuzhouFujian350001China
| | - Fei Gao
- Department of Anesthesiology/Critical Care MedicineFuzhou University Affiliated Provincial HospitalSchool of MedicineFuzhou UniversityShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial HospitalFuzhouFujian350001China
| | - Fu‐Shan Xue
- Department of Anesthesiology/Critical Care MedicineFuzhou University Affiliated Provincial HospitalSchool of MedicineFuzhou UniversityShengli Clinical Medical College of Fujian Medical UniversityFujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial HospitalFuzhouFujian350001China
| | - Xiaochun Zheng
- Department of AnesthesiologyFujian Provincial HospitalShengli Clinical Medical College of Fujian Medical University & Fujian Emergency Medical CenterFujian Provincial Key Laboratory of Emergency MedicineFujian Provincial Key Laboratory of Critical MedicineFujian Provincial Co‐constructed Laboratory of “Belt and Road,”FuzhouFujianChina
| |
Collapse
|
109
|
Gonelli CA, King HAD, Ko S, Fennessey CM, Iwamoto N, Mason RD, Heimann A, Flebbe DR, Todd JP, Foulds KE, Keele BF, Lifson JD, Koup RA, Roederer M. Antibody prophylaxis may mask subclinical SIV infections in macaques. Nature 2025; 639:205-213. [PMID: 39910294 PMCID: PMC11882457 DOI: 10.1038/s41586-024-08500-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/05/2024] [Indexed: 02/07/2025]
Abstract
Broadly neutralizing antibodies (bNAbs) show potential to prevent human immunodeficiency virus (HIV-1) infection in humans1. However, there are limited data on the antibody concentrations required to prevent infection. Clinical trials of bNAb prophylaxis have demonstrated partial efficacy2, but the sampling frequency typically does not allow precise timing of infection events and concurrent antibody levels. Here, using simian immunodeficiency virus (SIV) infection of rhesus macaques, we show that although potent bNAbs can delay the onset of acute viremia, subclinical infections occur while bNAb levels remain high. Serial SIV challenge of monkeys given partially and fully neutralizing bNAbs revealed that 'viral blips'-low and transient plasma viremia-often occur while serum bNAb concentrations are well above currently accepted protective levels. To understand the precise timing of the infections resulting in such blips, we performed plasma viral sequencing on monkeys that were serially challenged with genetically barcoded SIV after bNAb administration. These analyses showed that subclinical infections occurred in most animals that were given potent bNAb prophylaxis. These subclinical infections occurred while antibody concentrations were 2- to 400-fold higher than the levels required to prevent fully viremic breakthrough infection. This study demonstrates that immunoprophylaxis can mask subclinical infections, which may affect the interpretation of prophylactic HIV-1 bNAb clinical trials.
Collapse
Affiliation(s)
- Christopher A Gonelli
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hannah A D King
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - SungYoul Ko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nami Iwamoto
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rosemarie D Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ashley Heimann
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dillon R Flebbe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
110
|
Krinock DJ, Stephenson K, Irby D, Akmyradov C, Barker M, Waldrip Z, Burdine M, Wolf LL, Dassinger MS, Wyrick D. Intra-peritoneal Povidone-iodine Irrigation Decreases Abscesses in a Perforated Appendicitis Murine Model. J Pediatr Surg 2025; 60:162081. [PMID: 39657362 DOI: 10.1016/j.jpedsurg.2024.162081] [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: 08/22/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Children with perforated appendicitis frequently form post-operative intra-abdominal abscesses (IAA). Intra-peritoneal irrigation for prevention remains controversial. Using a perforated appendicitis murine model, we sought to determine the effect of intra-peritoneal irrigation on post-operative IAA and adhesion formation. METHODS A survival operation was performed where cecal ligation and puncture was used to simulate perforated appendicitis. After 72 h, mice underwent a second operation where a distal cecectomy and intra-peritoneal irrigation was performed. Mice were assigned to a no irrigation group or one of four irrigation groups: normal saline, povidone-iodine (PVI), tacrolimus, or PVI followed by tacrolimus. At 2-weeks or 2-months after the second survival operation, mice were euthanized and IAAs were counted, measured, and cultured. Intra-peritoneal adhesion severity was graded on a 4-point scale. Statistical analysis compared IAA numbers and adhesion grade between the irrigation groups. RESULTS In the 2-week cohort, prevalence of IAA was 78 % (n = 129). Type of irrigation solution was associated with abscess development (p < 0.001). Pairwise comparisons demonstrated PVI alone decreased abscess count. PVI irrigation caused more severe adhesions while tacrolimus decreased adhesions and adhesion grade was dependent on irrigation solution (p < 0.001). In the 2-month cohort, similar responses were observed with decreased abscess numbers in the PVI group (p = 0.006) but increased adhesion burden (p = 0.002). CONCLUSION Povidone-iodine irrigation decreases intra-abdominal abscess formation, but increases adhesion formation. LEVEL OF EVIDENCE: 1
Collapse
Affiliation(s)
- Derek J Krinock
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Krista Stephenson
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - David Irby
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chary Akmyradov
- Biostatistics Core, Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Melanie Barker
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Zachary Waldrip
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marie Burdine
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lindsey L Wolf
- Division of Pediatric Surgery, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Melvin S Dassinger
- Division of Pediatric Surgery, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Deidre Wyrick
- Division of Pediatric Surgery, Arkansas Children's Hospital, Little Rock, AR, USA
| |
Collapse
|
111
|
Papanikolaou GE, Kegels P, Gousios G, Kegels F, Peters L, Cremers N. Use of Supplemented Medical-Grade Honey to Treat Traumatic Skin Injuries in Geriatric Patients in a Home-Care Setting. Cureus 2025; 17:e80189. [PMID: 40196090 PMCID: PMC11973404 DOI: 10.7759/cureus.80189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Background Traumatic skin injuries (TSI) are more common to geriatric population due to reduced skin elasticity and increased gait instability. This is also associated with an altered wound-healing mechanism thus requires a cost-efficient and effective treatment. Therefore, the aim of the present study is to highlight the efficacy of medical-grade honey (MGH) supplemented with vitamins C and E for the conservative treatment of TSI in a home-care setting. Methodology The present multicenter retrospective case series study included 10 geriatric patients (four men and six women) who sustained TSI of various etiologies. The median age of the patients was 82.5 years (min-max: 65-90 years). Cardiovascular disease was the most frequent concomitant illness (50% of the patients). Previous treatments with povidone-iodine, alginate gel, or simple gauze for 5.5 days (median, min-max: 0-21 days) were ineffective. Treatment with a variety of supplemented MGH products, including ointment, wound gel, gauze, and foam, was commenced upon the initial patients' examination. Results By using MGH products, we achieved effective reduction of the inflammation, removal of the necrotic tissue, and formation of healthy granulation and epithelial tissue. MGH eliminated clinical signs of infection after 7.5 days (median, min-max: 5-35 days). Wounds were completely healed after 21 days (median, min-max: 14-56 days), without evidence of recurrence or complications, and with good functional and aesthetic outcomes. Conclusions Supplemented MGH-based products present high clinical efficacy for the treatment of TSI in older adults in a home-care setting, while demonstrating a safe and easy-to-use profile. Therefore, they can be proposed as an alternative or complementary therapeutic approach to conventional TSI therapies.
Collapse
Affiliation(s)
| | - Pieter Kegels
- Wound Care, Thuisverpleging Kegels, Sint-Gilles-Waas, BEL
| | | | - Filip Kegels
- Wound Care, Thuisverpleging Kegels, Sint-Gilles-Waas, BEL
| | - Linsey Peters
- Gynaecology and Obstetrics, Maastricht University Medical Centre, Maastricht, NLD
- Wound Care, Triticum Exploitatie BV, Maastricht, NLD
| | - Niels Cremers
- Gynaecology and Obstetrics, Maastricht University Medical Centre, Maastricht, NLD
- Wound Care, Triticum Exploitatie BV, Maastricht, NLD
| |
Collapse
|
112
|
Rubio-Casillas A, Redwan EM, Uversky VN. More antibodies are not always better: Fc effector functions play a critical role in SARS-CoV-2 infection and protection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:413-447. [PMID: 40246351 DOI: 10.1016/bs.pmbts.2025.02.001] [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: 04/19/2025]
Abstract
Traditional vaccinology has primarily focused on neutralizing antibody titers as the main correlate of vaccine efficacy, often overlooking the multifaceted roles of antibody Fc effector functions in orchestrating protective immune responses. Fc-mediated immune responses play a pivotal role in immune modulation and pathogen clearance. Emerging evidence from natural infections and vaccine studies highlights the critical contribution of Fc effector functions in determining the quality and durability of immunity. This work explores the limitations of current vaccine evaluation paradigms that prioritize neutralization over Fc effector mechanisms. It also describes findings from a study showing an unexpected role for SARS-CoV-2 anti-spike antibodies: both convalescent plasma and patient-derived monoclonal antibodies (mAbs) lead to maximum phagocytic capacity by monocytes at low concentrations, whereas at higher concentrations the phagocytic capacity was reduced. Given that the severity of COVID-19 disease and antibody titers are strongly positively correlated, this work challenges the paradigm that high antibodies offer better protection against severe disease. It is proposed that humoral and cellular responses elicited by vaccination should never be higher than those produced by natural infection. By integrating antibody Fc effector functions into vaccine development, a paradigm shift is proposed that emphasizes synergic antibody responses. Such an approach could transform vaccine efficacy assessment, enhance protection against dangerous pathogens, and drive innovation in vaccine design.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
113
|
Suchithra KV, Hameed A, Rekha PD, Stothard P, Arun AB. A novel Kayvirus species phage RuSa1 removes biofilm and lyses multiple clinical strains of methicillin resistant Staphylococcus aureus. Sci Rep 2025; 15:7358. [PMID: 40025202 PMCID: PMC11873149 DOI: 10.1038/s41598-025-92032-6] [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: 06/22/2024] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) infection is one of the global healthcare concerns. Here, we report the phenotypic and genotypic characterization of a novel multi-host Staphylococcus phage RuSa1, isolated from wastewater samples derived from a spotted sambar deer (Rusa unicolor) enclosure located at Mangalore, India. Clinical MRSA strains (n = 18) susceptible to RuSa1 were genetically and phenotypically diverse as determined by DNA fingerprinting and in vitro culture assays. RuSa1 displayed a latent period and burst size of 10 min and 50 PFU, respectively, and exhibited efficient biofilm removal activities against S. aureus ATCC BAA-44. The phage exhibited moderate UV stability (3 min) and high titre at 4-37 °C and pH 5‒9. RuSa1 possessed a linear double-stranded genomic DNA with a length of 140 kb. The genome contained 30.18% GC composition and shared 82.0‒94.9% sequence similarity with eleven authentic species of Kayvirus recognized by the International Committee on Taxonomy of Viruses based on VIRIDIC analysis. RuSa1 established distinct phyletic lineage in the maximum likelihood phylogenetic analysis of DNA encoding structural proteins and lacked genes that confer lysogeny. Based on the genotypic, phylogenetic and phenotypic data, RuSa1 is proposed to be a lytic phage and a new species of Kayvirus with a potential therapeutic ability against staphylococcal infections.
Collapse
Affiliation(s)
- Kokkarambath Vannadil Suchithra
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018, India
| | - Asif Hameed
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018, India.
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018, India
| | - Paul Stothard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Ananthapadmanabha Bhagwath Arun
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018, India.
- Yenepoya Institute of Arts, Science, Commerce and Management, Kulur, Mangalore, 575013, India.
| |
Collapse
|
114
|
Araiza-Atanacio I, Sáez-de-Ocariz M, Orozco-Covarrubias L. Transfusion-Associated Graft-Versus-Host Disease in Pediatric Patients: Clinical Features and Outcomes. Pediatr Dermatol 2025; 42:267-272. [PMID: 39581878 DOI: 10.1111/pde.15812] [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: 06/13/2023] [Revised: 09/06/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Transfusion-associated graft-versus-host disease (TA-GVHD) is a rare, usually fatal complication of blood transfusion. OBJECTIVE To describe the characteristics of TA-GVHD in children. METHODS The clinical records of pediatric patients diagnosed with TA-GVHD between January 2007 and December 2021 were reviewed. RESULTS We analyzed 94 clinical records of pediatric patients (0-18 years) with a diagnosis of GVHD, of whom 6 (6.38%) were associated with TA-GVHD; both genders being equally affected. The median age at diagnosis of the underlying disorder was 9 years, 1 month (range 2 months-15 years, 3 months); the median age at diagnosis of TA-GVHD was 9 years, 5.5 months (range 1 year, 7 months-15 years, 7 months). There were 27 grafts; 8 were irradiated and 12 were filtered. All patients presented with Stage 3 cutaneous TA-GVHD and histopathological Grade 2. All cases corresponded to classic acute TA-GVHD with global clinical Grade I-II. Mortality rate was 67%. The median follow-up of our patients was 2 years and 2 months (range 4 months-3 years, 4 months). CONCLUSIONS TA-GVHD, although less frequently encountered than in previous years, is still a concern. Early suspicion of TA-GVHD is mandatory due to its high mortality rates and rapid progression. Prevention by using irradiated blood products is the sole effective measure against this condition. Failing to identify and preempt TA-GVHD not only jeopardizes patient survival but also underscores the critical importance of vigilant monitoring and proactive intervention in at-risk patients.
Collapse
|
115
|
Wang Y, Lindsley K, Bleak TC, Jiudice S, Uyei J, Gu Y, Wang Y, Timbrook TT, Balada-Llasat JM. Performance of molecular tests for diagnosis of bloodstream infections in the clinical setting: a systematic literature review and meta-analysis. Clin Microbiol Infect 2025; 31:360-372. [PMID: 39672467 DOI: 10.1016/j.cmi.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Rapid identification of bloodstream pathogens and associated antimicrobial resistance (AMR) profiles by molecular tests from positive blood cultures (PBCs) have the potential to improve patient management and clinical outcomes. OBJECTIVES A systematic review and meta-analysis were conducted to evaluate diagnostic test accuracy (DTA) of molecular tests from PBCs for detecting pathogens and AMR in the clinical setting. METHODS . DATA SOURCES Medline, Embase, Cochrane, conference proceedings, and study bibliographies were searched. STUDY ELIGIBILITY CRITERIA Studies evaluating DTA of commercially available molecular tests vs. traditional phenotypic identification and susceptibility testing methods in patients with PBCs were eligible. PARTICIPANTS Patients with PBCs. TESTS Commercially available molecular tests. REFERENCE STANDARD Traditional phenotypic identification and susceptibility testing methods (standard of care, SOC). ASSESSMENT OF RISK OF BIAS Study quality was assessed using Quality Assessment of Diagnostic Accuracy Studies-2. METHODS OF DATA SYNTHESIS Summary DTA outcomes were estimated using bivariate random-effects models for gram-negative bacteria (GNB), gram-positive bacteria (GPB), yeast, GNB-AMR, GPB-AMR, and specific targets when reported by ≥ 2 studies (PROSPERO CRD42023488057). RESULTS Seventy-four studies including 24 590 samples were analysed, most of which had a low risk of bias. When compared with SOC, molecular tests showed 92-99% sensitivity, 99-100% specificity, 99-100% positive predictive value, and 97-100% negative predictive value for identifying total GNB (43 studies), GPB (38 studies), yeast (24 studies), GNB-AMR (35 studies), and GPB-AMR (39 studies). For individual pathogen targets, 93-100% sensitivity, 98-100% specificity, 86-100% positive predictive value, and 99-100% negative predictive value were estimated. Five of seven AMR genes had 91-99% sensitivity and 99-100% specificity. Sensitivity was lower for IMP (four studies; 62%; 95% CI, 34-83%) and VIM (four studies; 70%; 95% CI, 38-90%) carbapenemases, where genes were not detected or were not harboured in Pseudomonas aeruginosa (i.e. low prevalence). Performance of molecular tests in detecting AMR was generally comparable when grouped by geographical region (Europe, North America, and East Asia). DISCUSSION High DTA support the use of molecular tests in identifying a broad panel of pathogens and detecting AMR in GNB and GPB.
Collapse
Affiliation(s)
- Yu Wang
- IQVIA, Inc., Durham, NC, USA
| | | | - Tammy C Bleak
- Global Medical Affairs, bioMérieux, Salt Lake City, UT, USA
| | - Sarah Jiudice
- Global Medical Affairs, bioMérieux, Salt Lake City, UT, USA
| | | | | | - Yi Wang
- IQVIA, Inc., Durham, NC, USA
| | - Tristan T Timbrook
- Global Medical Affairs, bioMérieux, Salt Lake City, UT, USA; University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | | |
Collapse
|
116
|
Turcu-Stiolica A, Boicea AR, Nechita F, Surugiu R, Dumitra GG, Oancea CN, Pîrlog MC. Development and validation of a questionnaire to measure COVID-19 vaccine hesitancy within the Romanian industrial laborers. Front Public Health 2025; 13:1482778. [PMID: 40084211 PMCID: PMC11903443 DOI: 10.3389/fpubh.2025.1482778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
The identification and quantification of sources of vaccine hesitancy among industrial workers in Romania have become crucial for developing effective strategies to facilitate the vaccination process. Our study included employees, both with and without comorbidities, who work in industrial companies. The goal was to develop a scale to assess COVID-19 vaccine hesitancy in Romania. This proposed scale has been designated as the Romanian COVID-19 Vaccine Hesitancy (RO-CVH) scale. The survey encompassed both the demographic characteristics of the respondents and questions related to their perceptions of COVID-19 vaccination. A three-stage process was used to develop the RO-CVH which includes (1) item generation; (2) item-refinement (pilot testing, exploratory factor analysis); and (3) scale validation. The fifteen items loaded onto three factors using exploratory factor analysis, explaining 63% of the total variance. The three factors were labelled as "Confidence in information regarding the COVID-19 vaccine," "Safety and efficacy of the COVID-19 vaccine," and "COVID-19 vaccination as a means of controlling the population." The content validity of the scale was established, and it will be utilized to comprehend the behavior of industrial workers in Romania during similar future outbreaks, particularly regarding the acceptance of mitigatory vaccines. Based on the insights from this scale, future interventions could be designed to reduce vaccine hesitancy.
Collapse
Affiliation(s)
- Adina Turcu-Stiolica
- Department of Pharmacoeconomics, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Ancuta Ramona Boicea
- Department of Occupational Medicine, University of Medicine, and Pharmacy of Craiova, Craiova, Romania
| | - Florina Nechita
- Department of Medical Psychology, University of Medicine, and Pharmacy of Craiova, Craiova, Romania
| | - Roxana Surugiu
- Department of Biochemistry, University of Medicine, and Pharmacy of Craiova, Craiova, Romania
| | | | - Carmen Nicoleta Oancea
- Department of Biochemistry, University of Medicine, and Pharmacy of Craiova, Craiova, Romania
| | - Mihail Cristian Pîrlog
- Department of Medical Sociology, University of Medicine, and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
117
|
Vázquez E, de Gregorio O, Álvarez C, Soriano V, Corral O, Ortega-de la Puente A, de la Cruz-Echeandía M, Blanco-Valencia XP, Royuela A, Martín-Portugués M, Esteban-Sampedro J, Moreno-Torres V. Impact of immunosuppression on Listeria monocytogenes infection in Spain. Pathog Glob Health 2025:1-7. [PMID: 40015696 DOI: 10.1080/20477724.2025.2472300] [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: 11/15/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Immunosuppression (IS) determines a higher risk of disease severity from Listeria monocytogenes (LM) infection. METHODS We examined the epidemiology of IS in all patients hospitalized with LM in Spain from 2000 to 2021 in the National Registry of Hospital Discharges. IS was defined by liver disease (LD), diabetes mellitus (DM), chronic kidney disease (CKD), solid organ transplantation (SOT), bone marrow transplantation (BMT), primary immunodeficiencies (ID), systemic autoimmune diseases (SAD), solid organ neoplasms (SON), and hematological neoplasms (HN). RESULTS Among 8,152 admissions with LM, 48% were IS. There was an increase from 39.5% to 60% during the study period, mainly driven by rises in DM (from 12.6% to 26.2%), SON (from 9.9% to 17.5%), CKD (from 4.4% to 16.3%), HN (from 6.6% to 13.4%), and LD (from 4.9% to 6.6%) (p < 0.001 for all trends). IS fatality rate was higher than in non-IS (22.4% vs 11.3%; OR = 2.09). The proportion of LM patients with IS among LM in-hospital deaths increased from 57.1% in 2000 to 67.95% in 2021 (p < 0.001). Fatality risk differed according to baseline IS condition: LD (OR = 2.42), CKD (OR = 1.49), SON (OR 3.01) and HN (OR 1.45). CONCLUSIONS The prevalence of IS among patients hospitalized with LM in Spain has risen over the past two decades, with a growing impact on fatality rates. These findings should prompt further efforts to prevent and manage properly LM infection.
Collapse
Affiliation(s)
- Elena Vázquez
- UNIR Health Sciences School and Medical Center, Madrid, Spain
| | | | - Carmen Álvarez
- UNIR Health Sciences School and Medical Center, Madrid, Spain
| | - Vicente Soriano
- UNIR Health Sciences School and Medical Center, Madrid, Spain
| | - Octavio Corral
- UNIR Health Sciences School and Medical Center, Madrid, Spain
| | | | | | | | - Ana Royuela
- Biostatistics Unit, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain
- Center for Biomedical Research in Epidemiology and Public Health Network, CIBERESP, Madrid, Spain
| | - Mario Martín-Portugués
- Internal Medicine Department, Health Research Institute Puerta de Hierro-Segovia de Arana (IDIPHIM), Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Jorge Esteban-Sampedro
- Internal Medicine Department, Health Research Institute Puerta de Hierro-Segovia de Arana (IDIPHIM), Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Víctor Moreno-Torres
- UNIR Health Sciences School and Medical Center, Madrid, Spain
- Internal Medicine Department, Health Research Institute Puerta de Hierro-Segovia de Arana (IDIPHIM), Hospital Universitario Puerta de Hierro, Madrid, Spain
| |
Collapse
|
118
|
Happe M, Lynch RM, Fichtenbaum CJ, Heath SL, Koletar SL, Landovitz RJ, Presti RM, Santana-Bagur JL, Tressler RL, Holman LA, Novik L, Roa JC, Rothwell RS, Strom L, Wang J, Hu Z, Conan-Cibotti M, Bhatnagar AM, Dwyer B, Ko SH, Belinky F, Namboodiri AM, Pandey JP, Carroll R, Basappa M, Serebryannyy L, Narpala SR, Lin BC, McDermott AB, Boritz EA, Capparelli EV, Coates EE, Koup RA, Ledgerwood JE, Mascola JR, Chen GL, Tebas P, the VRC 607/A5378 Study Team. Virologic effects of broadly neutralizing antibodies VRC01LS and VRC07-523LS on chronic HIV-1 infection. JCI Insight 2025; 10:e181496. [PMID: 39989458 PMCID: PMC11949028 DOI: 10.1172/jci.insight.181496] [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: 05/14/2024] [Accepted: 01/13/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUNDHIV-1-specific broadly neutralizing monoclonal antibodies (bNAbs) have emerged as promising interventions with the potential to effectively treat and prevent HIV-1 infections. We conducted a phase I clinical trial evaluating the potent CD4-binding site-specific (CD4bs-specific) bNAbs VRC01LS and VRC07-523LS in people with HIV-1 (PWH) not receiving antiretroviral therapy (ART).METHODSParticipants received a single intravenous 40 mg/kg dose of either VRC01LS (n = 7) or VRC07-523LS (n = 9) and did not initiate ART for a minimum of 14 days. The primary study objective was to evaluate safety and tolerability; the secondary study objectives were to evaluate pharmacokinetics (PK) and the impact of administered bNAbs on viral loads (VL) and CD4+ T cell counts in the absence of ART.RESULTSThis trial enrolled 16 PWH aged 20 to 57 years. Both bNAbs were safe and well tolerated. Mild local reactogenicity was only reported in participants who received VRC07-523LS, while both bNAbs were associated with mild systemic symptoms. Maximum serum concentrations (Cmax) following VRC01LS or VRC07-523LS were 1,566 ± 316 and 1,295 ± 376 μg/mL, respectively. VRC07-523LS administration significantly decreased VL in 8 out of 9 participants, with an average decline of 1.7 ± 0.8 log10 copies/mL within 14 days after administration. In contrast, VRC01LS administration resulted in a smaller average decline (0.8 ± 0.8 log10 copies/mL), and 3 out of 7 participants showedno change in VL. Postinfusion maximum decline in VL correlated with post hoc baseline in vitro viral susceptibility results for both bNAbs.CONCLUSIONThe results of this trial support inclusion of potent CD4bs-specific bNAbs, such as VRC07-523LS, into next-generation treatment regimens for HIV-1.TRIAL REGISTRATIONClinicalTrials.gov NCT02840474.FUNDINGNational Institute of Allergy and Infectious Diseases (NIAID)/NIH (grants UM1 AI068634, UM1 AI068636, UM1 AI106701, UM1AI069424, UM1AI069501, UM1AI69415, UM1AI069534, UM1AI69494); the Intramural Research Program of the NIAID/NIH; National Center for Advancing Translational Sciences/NIH (grants UM1TR004548, UL1TR001881, and UL1TR001878); and the National Cancer Institute/NIH (contract 75N91019D00024).
Collapse
Affiliation(s)
- Myra Happe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rebecca M. Lynch
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | | | - Sonya L. Heath
- Division of Infectious Diseases, University of Alabama, Birmingham, Alabama, USA
| | - Susan L. Koletar
- Division of Infectious Diseases, The Ohio State University, Columbus, Ohio, USA
| | - Raphael J. Landovitz
- Division of Infectious Diseases, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Rachel M. Presti
- Division of Infectious Diseases, Washington University, St. Louis, Missouri, USA
| | | | - Randall L. Tressler
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - LaSonji A. Holman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Laura Novik
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Jhoanna C. Roa
- AIDS Network Coordinating Center, DLH Corporation, Bethesda, Maryland, USA
| | - Ro Shauna Rothwell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Larisa Strom
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Jing Wang
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Zonghui Hu
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Michelle Conan-Cibotti
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Anjali M. Bhatnagar
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Bridget Dwyer
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Sung Hee Ko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Frida Belinky
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Aryan M. Namboodiri
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Janardan P. Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robin Carroll
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Leonid Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sandeep R. Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Bob C. Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Eli A. Boritz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Edmund V. Capparelli
- School of Medicine and Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Emily E. Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Julie E. Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Grace L. Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Pablo Tebas
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
119
|
Lejeune A, Zhou C, Ercelen D, Putzel G, Yao X, Guy AR, Pawline M, Podkowik M, Pironti A, Torres VJ, Shopsin B, Cadwell K. Sex-dependent gastrointestinal colonization resistance to MRSA is microbiota and Th17 dependent. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603994. [PMID: 39763855 PMCID: PMC11702559 DOI: 10.1101/2024.07.17.603994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Gastrointestinal (GI) colonization by methicillin-resistant Staphylococcus aureus (MRSA) is associated with a high risk of transmission and invasive disease in vulnerable populations. The immune and microbial factors that permit GI colonization remain unknown. Male sex is correlated with enhanced Staphylococcus aureus nasal carriage, skin and soft tissue infections, and bacterial sepsis. Here, we established a mouse model of sexual dimorphism during GI colonization by MRSA. Our results show that in contrast to male mice that were susceptible to persistent colonization, female mice rapidly cleared MRSA from the GI tract following oral inoculation in a manner dependent on the gut microbiota. This colonization resistance displayed by female mice was mediated by an increase in IL-17A+ CD4+ T cells (Th17) and dependent on neutrophils. Ovariectomy of female mice increased MRSA burden, but gonadal female mice that have the Y chromosome retained enhanced Th17 responses and colonization resistance. Our study reveals a novel intersection between sex and gut microbiota underlying colonization resistance against a major widespread pathogen.
Collapse
|
120
|
Soltani S, Moradinazar M, Karamimatin B, Gouya MM, Zahraei SM, Moradi G, Chehri O, Soofi M, Shadmani FK, Kalantari M, Najafi F. Monitoring and active surveillance of adverse events following the booster dose of AZD1222 vaccine in people vaccinated with Sinopharm BBIBP-CorV: a cohort study. BMC Public Health 2025; 25:650. [PMID: 39962455 PMCID: PMC11834735 DOI: 10.1186/s12889-025-21805-5] [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: 05/10/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Given the limited evidence on the side effects of AZD1222 booster in individuals previously vaccinated with Sinopharm BBIBP-CorV, we conducted a cohort event monitoring study to capture adverse events in those receiving AZD1222 booster doses (first or second) after Sinopharm BBIBP-CorV vaccination in Iran. METHODS This active COVID-19 vaccine safety surveillance study was conducted in vaccination centers across 12 provinces in Iran. The study population included individuals who had received two doses of Sinopharm BBIBP-CorV and either a first or second AZD1222 booster dose. Each participant was followed for 13 weeks after their booster dose. RESULTS A total of 28,742 participants were included, with 13,428 females (46.72%) and 15,314 males (53.28%). The average age was 43.25 ± 14.90 years. Of the total, 82.46% were recruited retrospectively through vaccination registration. Among 16,944 individuals with available polymerase chain reaction test results, 118 tested positive for COVID-19 between 14 and 90 days post-booster. There were 123 hospital admissions, with 18 cases showing a higher likelihood of being associated with adverse events following immunization (AEFIs) or serious adverse events (SAEs). After excluding deaths unrelated to vaccination, four deaths were potentially linked to AEFIs or SAEs. The cumulative incidence rates for hospital admissions and deaths were 106.40 and 13.90 per 100,000 individuals, respectively. CONCLUSION The study indicates that AZD1222 booster doses are safe for individuals previously vaccinated with Sinopharm BBIBP-CorV, with rare adverse events and an acceptable safety profile. This supports the use of heterologous booster strategies, though ongoing monitoring and improved pharmacovigilance are essential to detect any potential long-term effects.
Collapse
Affiliation(s)
- Shahin Soltani
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Moradinazar
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behzad Karamimatin
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Mehdi Gouya
- Iranian Center for Communicable Diseases Control, Ministry of Health & Medical Education, Tehran, Iran
| | - Sayed Mohsen Zahraei
- Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghobad Moradi
- Social Determinant of the Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Omid Chehri
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Moslem Soofi
- Social Development and Health Promotion Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khosravi Shadmani
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahsa Kalantari
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farid Najafi
- Research Center for Environmental Determinants of Health, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
121
|
Durham K, Atagozli T, Elliott DE, Ince MN. Laboratory Tests in Inflammatory Bowel Disease: An Evidence-Based Approach to Daily Practice. Biomedicines 2025; 13:491. [PMID: 40002904 PMCID: PMC11852734 DOI: 10.3390/biomedicines13020491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) comprise a group of chronic gastrointestinal disorders characterized by periods of relapse and remission. The mainstay of treatment is medical, involving medications such as steroids, immune modulators, monoclonal antibodies (categorized as biologics), and small molecules. These medications can provide profound therapeutic benefits, but they can also cause severe and irreversible toxicities. Clinicians may utilize laboratory tests in the diagnosis and management of IBD including assessment of disease activity, monitoring medication response or toxicity, surveillance of infectious complications, and detection of nutritional deficiencies. Routine use of laboratory tests may help clinicians avoid reactivation of life-threatening infections such as tuberculosis or hepatitis B virus upon initiation of immune suppressive therapy. They can also be used to detect vitamin deficiencies such as B12 deficiency, which has the potential to cause irreversible neurologic damage. While some laboratory tests constitute established practices, the utility of newer tests such therapeutic drug monitoring (TDM) in the era of biologics is an evolving topic. Although clinical assessment with imaging, endoscopic, and histopathological examination is standard practice, laboratory tests serve as valuable adjuncts. We aim to explore the broad range of laboratory tests available to clinicians and to summarize their application in the current management of IBD in daily clinical practice, with special attention to updates in therapeutic drug monitoring.
Collapse
Affiliation(s)
- Katelin Durham
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242, USA; (K.D.); (D.E.E.)
- Iowa City Veterans Affairs Medical Center, 601 Highway 6 W, Iowa City, IA 52246, USA
| | - Tyler Atagozli
- Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA 52242, USA;
| | - David E. Elliott
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242, USA; (K.D.); (D.E.E.)
- Iowa City Veterans Affairs Medical Center, 601 Highway 6 W, Iowa City, IA 52246, USA
| | - M. Nedim Ince
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242, USA; (K.D.); (D.E.E.)
- Iowa City Veterans Affairs Medical Center, 601 Highway 6 W, Iowa City, IA 52246, USA
| |
Collapse
|
122
|
Tamagne M, Khelfa M, Many S, Neyrinck-Leglantier D, Delorme AS, Pinheiro MK, Andrieu M, Cleophax S, Pirenne F, Vingert B. Interactions with and activation of immune cells by CD41a + extracellular vesicles. Front Immunol 2025; 16:1509078. [PMID: 40028321 PMCID: PMC11868057 DOI: 10.3389/fimmu.2025.1509078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction The immunological profiles of CD4+ T lymphocytes (TLs) from patients with hematological malignancies differ between patients who have and have not received transfusions. There may be several reasons for these differences, including the presence of extracellular vesicles (EVs) derived from plasma membrane budding and present in the platelet concentrates. Indeed, EVs can modulate the immune system through interactions with many immune cells, but the underlying mechanisms remain incompletely understood. Methods We therefore investigated how interactions with CD41a+ EVs cause immune cells to change phenotype and function. CD41a+ EVs were cultured with TLs, B lymphocytes, and monocytes. Given the potential involvement of monocytes in leukemia progression, we performed a new original multi-omics study to confirm the protein changes and gene activation observed following interaction with CD41a+ EVs. Results The CD41a+ EVs had immunomodulatory effects on all these cell types but this effect depended on the numbers of EVs. CD4+ TLs required large numbers of CD41a+ EVs for activation, whereas monocytes were the most sensitive. With the new multi-omics technique, we confirmed the direct effects of CD41a+ EVs on protein phenotype and gene activation. Conclusion Transfusion EVs should be considered during the immunological follow-up of patients after transfusion to detect immunological effects on malignant hemopathies, and during the development of new immunotherapies.
Collapse
Affiliation(s)
- Marie Tamagne
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| | - Mehdi Khelfa
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| | - Souganya Many
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | | | - Adèle Silane Delorme
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| | - Marion Klea Pinheiro
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| | - Muriel Andrieu
- Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique (CNRS) UMR8104, Université Paris-Cité, Paris, France
| | | | - France Pirenne
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| | - Benoît Vingert
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Etablissement Français du Sang, Ivry sur Seine, France
| |
Collapse
|
123
|
Ahmann M, Compton J, Pottinger J, Uhlenhopp R, Ward M, Haleem A, Willey M, Schweizer M, Herwaldt L. Staphylococcus aureus colonization and surgical site infections among patients undergoing surgical fixation for acute fractures. Infect Control Hosp Epidemiol 2025; 46:1-7. [PMID: 39949120 PMCID: PMC12015622 DOI: 10.1017/ice.2024.233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 04/25/2025]
Abstract
OBJECTIVES To identify risk factors for methicillin-susceptible (MSSA) and methicillin-resistant S. aureus (MRSA) nasal carriage and surgical site infection (SSI) among patients undergoing fracture fixation procedures who were included in a quality improvement protocol involving screening patients for S. aureus nasal carriage and treating carriers with intranasal mupirocin and chlorhexidine bathing. DESIGN Retrospective cohort study. SETTING Level 1 trauma center. PARTICIPANTS 1,254 adults who underwent operative fixation of 1,298 extremity or pelvis fractures between 8/1/2014 - 7/31/2017. METHODS We calculated rates of S. aureus nasal carriage and SSI. We used multivariable stepwise logistic regression and selected the final models based on Akaike information criterion. RESULTS Of the 1,040 screened first procedures, 262 (25.19%) were performed on S. aureus nasal carriers: 211 (20.29%) on MSSA carriers and 51 (4.90%) on MRSA carriers. Long-term care facility residence (odds ratio [OR] 3.38; 95% confidence interval [CI] 1.17-9.76) was associated with MRSA nasal carriage. After adjusting for statistically and clinically significant variables, MRSA carriage was significantly associated with any SSI (OR 4.58; 95% CI 1.63-12.88), S. aureus SSI (OR 10.11; 95% CI 3.25-31.42), and MRSA SSI (OR 27.25; 95% CI 5.33-139.24), whereas MSSA carriage was not. Among S. aureus carriers, any chlorhexidine use was documented for 232 (88.55%), and any intranasal mupirocin was documented for 85 (40.28%) MSSA carriers and 33 (64.71%) MRSA carriers. CONCLUSIONS MRSA carriage was associated with a significant risk of SSI after operative fracture fixation. Many carriers did not undergo decolonization, suggesting that a simplified decolonization protocol is needed.
Collapse
Affiliation(s)
- Megan Ahmann
- State Hygienic Laboratory at the University of Iowa, Coralville, IA, USA
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Jocelyn Compton
- Department of Orthopedics and Rehabilitation, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Orthopedic + Fracture Specialists, Portland, OR, USA
| | - Jean Pottinger
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Richard Uhlenhopp
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Melissa Ward
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Ambar Haleem
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Michael Willey
- Department of Orthopedics and Rehabilitation, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Marin Schweizer
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Loreen Herwaldt
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
124
|
Morales-Pacheco M, Valenzuela-Mayen M, Gonzalez-Alatriste AM, Mendoza-Almanza G, Cortés-Ramírez SA, Losada-García A, Rodríguez-Martínez G, González-Ramírez I, Maldonado-Lagunas V, Vazquez-Santillan K, González-Covarrubias V, Pérez-Plasencia C, Rodríguez-Dorantes M. The role of platelets in cancer: from their influence on tumor progression to their potential use in liquid biopsy. Biomark Res 2025; 13:27. [PMID: 39934930 DOI: 10.1186/s40364-025-00742-w] [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: 10/11/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Platelets, anucleate blood cells essential for hemostasis, are increasingly recognized for their role in cancer, challenging the traditional notion of their sole involvement in blood coagulation. It has been demonstrated that platelets establish bidirectional communication with tumor cells, contributing to tumor progression and metastasis through diverse molecular mechanisms such as modulation of proliferation, angiogenesis, epithelial-mesenchymal transition, resistance to anoikis, immune evasion, extravasation, chemoresistance, among other processes. Reciprocally, cancer significantly alters platelets in their count and composition, including mRNA, non-coding RNA, proteins, and lipids, product of both internal synthesis and the uptake of tumor-derived molecules. This phenomenon gives rise to tumor-educated platelets (TEPs), which are emerging as promising tools for the development of liquid biopsies. In this review, we provide a detailed overview of the dynamic roles of platelets in tumor development and progression as well as their use in diagnosis and prognosis. We also provide our view on current limitations, challenges and future research areas, including the need to design more efficient strategies for their isolation and analysis, as well as the validation of their sensitivity and specificity through large-scale and rigorous clinical trials. This research will not only enable the evaluation of their clinical viability but could also open new opportunities to enhance diagnostic accuracy and develop personalized treatments in oncology.
Collapse
Affiliation(s)
- Miguel Morales-Pacheco
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | - Miguel Valenzuela-Mayen
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | | | - Gretel Mendoza-Almanza
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Sergio A Cortés-Ramírez
- Department of Pharmacology and Toxicology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Alberto Losada-García
- Department of Pharmacology and Toxicology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Griselda Rodríguez-Martínez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
- Laboratorio de Investigación en Patógenos Respiratorios y Producción de Biológicos, Hospital Infantil de México Federico Gómez, Mexico City, 14610, Mexico
| | - Imelda González-Ramírez
- Departamento de Atención a La Salud, Universidad Autónoma Metropolitana Xochimilco, Mexico City, 14610, Mexico
| | - Vilma Maldonado-Lagunas
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Karla Vazquez-Santillan
- Laboratorio de Innovación en Medicina de Precisión, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Vanessa González-Covarrubias
- Laboratorio de Farmacogenómica, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, FES-Iztacala, Universidad Nacional Autónoma de México (UNAM), Iztacala, Tlalnepantla, 54090, Mexico
| | | |
Collapse
|
125
|
Starikova EA, Mammedova JT, Rubinstein AA, Sokolov AV, Kudryavtsev IV. Activation of the Coagulation Cascade as a Universal Danger Sign. Curr Issues Mol Biol 2025; 47:108. [PMID: 39996829 PMCID: PMC11854423 DOI: 10.3390/cimb47020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Hemostasis is a mechanism that stops bleeding from an injured vessel, involves multiple interlinked steps, culminating in the formation of a "clot" sealing the damaged area. Moreover, it has long been recognized that inflammation also provokes the activation of the coagulation system. However, there has been an increasing amount of evidence revealing the immune function of the hemostasis system. This review collects and analyzes the results of the experimental studies and data from clinical observations confirming the inflammatory function of hemostasis. Here, we summarize the latest knowledge of the pathways in immune system activation under the influence of coagulation factors. The data analyzed allow us to consider the components of hemostasis as receptors recognizing «foreign» or damaged «self» or/and as «self» damage signals that initiate and reinforce inflammation and affect the direction of the adaptive immune response. To sum up, the findings collected in the review allow us to classify the coagulation factors, such as Damage-Associated Molecular Patterns that break down the conventional concepts of the coagulation system.
Collapse
Affiliation(s)
- Eleonora A. Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Department of Microbiology and Virology, Institute of Medical Education Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 Saint Petersburg, Russia
| | - Jennet T. Mammedova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Department of Molecular Biotechnology, Chemical and Biotechnology Faculty, Saint Petersburg State Institute of Technology, Moskovski Ave., 26, 190013 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
| | - Alexey V. Sokolov
- Laboratory of Systemic Virology, Department of Molecular Biology of Viruses, Smorodintsev Research Institute of Influenza, 15/17, Prof. Popova Str., 197376 Saint Petersburg, Russia;
| | - Igor V. Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| |
Collapse
|
126
|
Fang J, Wang Z, Shen Y, Wu X, Fang H, Sun X, Yu T, Zhang Q. Case report: The value of early application of mNGS technology in the diagnosis and treatment of severe Legionnaires' disease: reports of two cases with different outcomes. Front Med (Lausanne) 2025; 12:1501192. [PMID: 39975683 PMCID: PMC11835848 DOI: 10.3389/fmed.2025.1501192] [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: 09/24/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025] Open
Abstract
Background Legionnaires' disease has a high clinical mortality rate, and early diagnosis and treatment are critical. Increasing evidence shows that metagenomic next-generation sequencing (mNGS) has excellent potential for the early identification of pathogens. To help clinicians better recognize Legionnaires' disease in its early stage and to illustrate the diagnostic value of mNGS technology, we reviewed and summarized two cases of severe Legionnaires' disease. Methods and analysis We selected two patients with severe Legionnaires' disease who were admitted to our department in recent years. We discuss experience with them and the shortcomings in their treatment by summarizing their medical history, disease evolution, tests, and diagnostic and therapeutic processes. Results In both patients, the diagnosis of Legionnaires' disease was confirmed through analysis of the bronchoalveolar lavage fluid (BALF). The middle-aged male patient was cured and discharged due to early detection and diagnosis. The elderly immunocompromised patient died due to a delay in diagnosis. Conclusion This study highlights the importance of the early recognition and diagnosis of severe Legionnaires' disease and the advantages of mNGS in identifying the pathogen.
Collapse
Affiliation(s)
- Jianqing Fang
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Zhe Wang
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Yu Shen
- Department of Ultrasound Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Xuenong Wu
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Hao Fang
- Department of Intensive Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Xiaokui Sun
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ting Yu
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Qingqing Zhang
- Department of Respiratory and Critical Care Medicine, Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
127
|
Nowacki JS, Jones GS, D'Orazio SEF. Listeria monocytogenes use multiple mechanisms to disseminate from the intestinal lamina propria to the mesenteric lymph nodes. Microbiol Spectr 2025; 13:e0259524. [PMID: 39714174 PMCID: PMC11792513 DOI: 10.1128/spectrum.02595-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024] Open
Abstract
Listeria monocytogenes are facultative intracellular bacterial pathogens that cause foodborne disease in humans. The bacteria can use the surface protein InlA to invade intestinal epithelial cells or transcytose across M cells in the gut, but it is not well understood how the bacteria traffic from the underlying lamina propria to the draining mesenteric lymph nodes (MLN). Previous studies indicated that L. monocytogenes associated with both monocytes and dendritic cells in the intestinal lamina propria. We show here that CCR2-/- mice had a significant reduction in Ly6Chi monocytes in the MLN but no change in bacterial burden following foodborne infection; thus, dissemination of L. monocytogenes associated with monocytes is not required for colonization of the MLN. To block CCR7-mediated trafficking of dendritic cells from the lamina propria, we treated mice with anti-VEGFR3 antibody (clone AFL4) prior to and during infection but did not see a change in dendritic numbers in the MLN as had been previously reported with other anti-VEGFR3-specific antibodies. However, increasing the number of circulating dendritic cells by treating mice with rFlt3L resulted in a significant increase in L. monocytogenes in the lymph nodes that drain the small intestine and the spleen. Whole-mount fluorescent microscopy of lymphatic vessels following ligated loop infection revealed both free-floating L. monocytogenes and cell-associated bacteria within lymphatic vessels. Together, these results suggest that L. monocytogenes can use multiple, redundant mechanisms to disseminate from the gut tissue to the MLN. IMPORTANCE Consumption of the foodborne bacterial pathogen Listeria monocytogenes results in a wide spectrum of human disease from mild self-limiting gastroenteritis to life-threatening infections of the bloodstream, brain, and placenta. It is not well understood how the bacteria migrate from the intestines to the draining mesenteric lymph nodes, which are thought to serve as the last barrier to prevent systemic infections. Results presented here reveal multiple redundant mechanisms L. monocytogenes can use to disseminate from the ileum or colon to the mesenteric lymph nodes.
Collapse
Affiliation(s)
- Joshua S. Nowacki
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Grant S. Jones
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Sarah E. F. D'Orazio
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
128
|
Al Qureshah F, Le Pen J, de Weerd NA, Moncada-Velez M, Materna M, Lin DC, Milisavljevic B, Vianna F, Bizien L, Lorenzo L, Lecuit M, Pommier JD, Keles S, Ozcelik T, Pedraza-Sanchez S, de Prost N, El Zein L, Hammoud H, Ng LFP, Halwani R, Saheb Sharif-Askari N, Lau YL, Tam AR, Singh N, Bhattad S, Berkun Y, Chantratita W, Aguilar-López R, Shahrooei M, Abel L, Bastard P, Jouanguy E, Béziat V, Zhang P, Rice CM, Cobat A, Zhang SY, Hertzog PJ, Casanova JL, Zhang Q. A common form of dominant human IFNAR1 deficiency impairs IFN-α and -ω but not IFN-β-dependent immunity. J Exp Med 2025; 222:e20241413. [PMID: 39680367 DOI: 10.1084/jem.20241413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/13/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024] Open
Abstract
Autosomal recessive deficiency of the IFNAR1 or IFNAR2 chain of the human type I IFN receptor abolishes cellular responses to IFN-α, -β, and -ω, underlies severe viral diseases, and is globally very rare, except for IFNAR1 and IFNAR2 deficiency in Western Polynesia and the Arctic, respectively. We report 11 human IFNAR1 alleles, the products of which impair but do not abolish responses to IFN-α and -ω without affecting responses to IFN-β. Ten of these alleles are rare in all populations studied, but the remaining allele (P335del) is common in Southern China (minor allele frequency ≈2%). Cells heterozygous for these variants display a dominant phenotype in vitro with impaired responses to IFN-α and -ω, but not -β, and viral susceptibility. Negative dominance, rather than haploinsufficiency, accounts for this dominance. Patients heterozygous for these variants are prone to viral diseases, attesting to both the dominance of these variants clinically and the importance of IFN-α and -ω for protective immunity against some viruses.
Collapse
Affiliation(s)
- Fahd Al Qureshah
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology , Riyadh, Saudi Arabia
| | - Jérémie Le Pen
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Nicole A de Weerd
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Australia
| | - Marcela Moncada-Velez
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Daniel C Lin
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Université Paris Cité, Imagine Institute , Paris, France
| | - Baptiste Milisavljevic
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Fernanda Vianna
- Laboratório de Medicina Genômica Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Genetics and Molecular Biology, Federal University of Rio Grande do Sul , Porto Alegre, Brazil
- Graduate Program in Medicine, Medical Sciences, Federal University of Rio Grande do Sul , Porto Alegre, Brazil
- National Institute of Population Medical Genetics (INAGEMP) , Porto Alegre, Brazil
| | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Marc Lecuit
- Université Paris Cité, Imagine Institute , Paris, France
- Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, APHP, Institut Imagine, Paris, France
- Biology of Infection Unit, Institut Pasteur, Inserm U1117, Université Paris Cité, Paris, France
| | - Jean-David Pommier
- Biology of Infection Unit, Institut Pasteur, Inserm U1117, Université Paris Cité, Paris, France
| | - Sevgi Keles
- Division of Pediatric Allergy and Immunology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Tayfun Ozcelik
- Department of Molecular Biology and Genetics, Bilkent University, Bilkent-Ankara, Turkey
| | - Sigifredo Pedraza-Sanchez
- Unit of Biochemistry, National Institute for Medical Sciences and Nutrition Salvador Zubiran (INCMNSZ) , Mexico City, Mexico
| | - Nicolas de Prost
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP) , Paris, France
- Groupe de Recherche Clinique CARMAS, Faculté de Santé de Créteil, Université Paris Est Créteil , Créteil Cedex, France
- INSERM U955, Team "Viruses, Hepatology, Cancer" , Créteil, France
| | - Loubna El Zein
- Biology Department, Lebanese University, Beirut, Lebanon
| | | | - Lisa F P Ng
- A*STAR Infectious Disease Labs, Agency for Science, Technology and Research , Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University , Singapore, Singapore
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah , Sharjah, UAE
- Prince Abdullah Bin Khalid Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Yu Lung Lau
- Department of Pediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony R Tam
- Division of Infectious Diseases, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | | | | | - Yackov Berkun
- Department of Pediatrics, Hadassah-Hebrew University Medical Center, Mount Scopus and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wasun Chantratita
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Raúl Aguilar-López
- Department of Surgery, Maternal and Child Hospital, Social Security Institute of the State of Mexico and Municipalities (ISSEMYM), Toluca, Mexico
| | - Mohammad Shahrooei
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Dr. Shahrooei's Laboratory , Tehran, Iran
| | - Laurent Abel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Paul Bastard
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris , Paris, France
| | - Emmanuelle Jouanguy
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Vivien Béziat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Peng Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Aurélie Cobat
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Shen-Ying Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Clayton, Australia
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
- Howard Hughes Medical Institute , New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Qian Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute , Paris, France
| |
Collapse
|
129
|
Alam M, Islam MS, Jahan MI, Deb AS, Rahman A, Islam Z, Chowdhury AI, Islam KM, Hossain MZ, Ahmed D, Arifeen SE, Gurley ES, Rahman M. A novel virulent core genome multilocus sequence type CT 11424 of Listeria monocytogenes isolate causing stillbirth in Bangladesh. BMC Microbiol 2025; 25:61. [PMID: 39901076 PMCID: PMC11792674 DOI: 10.1186/s12866-024-03650-5] [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: 05/31/2024] [Accepted: 11/14/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Listeria monocytogenes is a foodborne pathogen that can lead to severe pregnancy outcomes. This study reports the clinical and genomic characteristics of a Listeria-mediated stillbirth identified in January 2022 through the Child Health and Mortality Prevention Surveillance (CHAMPS) project in Bangladesh. The Lm-BD-CHAMPS-01 isolate was recovered from the blood and cerebrospinal fluid (CSF) of a male stillborn. Maternal history, clinical, and demographic data were collected by the CHAMPS surveillance platform. An expert panel evaluated all reports to determine the role of L. monocytogenes infection in the causal chain of stillbirth. Genomic characterization included multilocus sequence typing (MLST), core genome MLST (cgMLST), serotyping, and the presence or absence of virulence genes. Genetic divergence and phylogenetic analyses were conducted to determine the relationship with other reported isolates globally. RESULTS The isolate Lm-BD-CHAMPS-01 was identified as a novel cgMLST CT11424. It belonged to ST 308, Serotype 4b, Clonal Complex 1, and Phylogenetic Lineage 1. Key L. monocytogenes virulence genes facilitating the crossing of the placental barrier, including full-length inlA, LIPI-1, and LIPI-3, were detected. The isolate was closely related to clinical L. monocytogenes isolates, as determined by GrapeTree based on cgMLST. SNP-based phylogenetic analysis found Lm-BD-CHAMPS-01 to be the most distant from other CC1 isolates in the database. Possible sources of infection included the consumption of contaminated raw vegetables or exposure to pigeons. CONCLUSIONS This is the first genome sequence of clinical L. monocytogenes from Bangladesh, which also caused stillbirth. Rural healthcare professionals should be aware of L. monocytogenes infection risks during pregnancy. Pregnant women should be counseled on the dangers of exposure to animals or birds and consumption of potentially contaminated raw food to prevent adverse pregnancy outcomes due to L. monocytogenes infection.
Collapse
Affiliation(s)
- Muntasir Alam
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh.
| | - Md Saiful Islam
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
- Division of Genomics & Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - M Ishrat Jahan
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
- Division of Genomics & Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Arpita Shyama Deb
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Afruna Rahman
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Zahidul Islam
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Atique Iqbal Chowdhury
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Kazi Munisul Islam
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Mohammad Zahid Hossain
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Dilruba Ahmed
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Shams El Arifeen
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| | - Emily S Gurley
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
- Infectious Disease Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Mustafizur Rahman
- International Centre for Diarrhoeal Disease Research, Infectious Diseases Division, 68 Shaheed Tajuddin Ahmed Sarani, (icddr,b), Dhaka, Mohakhali, 1212, Bangladesh
| |
Collapse
|
130
|
Anindya I, Sekartini R, Ariyanto IA, Wiguna T, Sari NP, Rahayu YS, Soebandrio A. Cytomegalovirus-Reactive IgG Correlates with Increased IL-6 and IL-1β Levels, Affecting Eating Behaviours and Tactile Sensitivity in Children with Autism. Biomedicines 2025; 13:338. [PMID: 40002751 PMCID: PMC11852405 DOI: 10.3390/biomedicines13020338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Elevated cytokine levels, including IL-6 and IL-1β, can contribute to persistent brain inflammation in children with autism and cytomegalovirus (CMV) infection, exacerbating autism-related behaviours and symptoms. This study evaluates the impact of CMV-induced cytokine increases on the eating behaviours and sensory profiles of children with autism. METHODS A cross-sectional design was employed, involving children aged two to five years (CMV-reactive IgG), with ASD (n= 98) and TD (n = 96). Serological tests using ELISA were conducted to measure IgG CMV, IL-6, and IL-1β biomarkers. Eating behaviours were evaluated using the BAMBI (Brief Autism Mealtime Behaviour Inventory), and sensory profiles were assessed using the SSP (Short Sensory Profile). Statistical analyses were performed using Spearman's rank and chi-square tests. RESULTS The results show that autism significantly affects children's eating behaviours and sensory profiles (p < 0.001), with notable differences found between the groups. Correlation analysis revealed a significant association between IgG CMV and IL-6 (p = 0.026) and IL-1β (p = 0.014) in the ASD group. Additionally, eating behaviours (food refusal and limited variety) in ASD correlated with IL-6 and IL-1β. Sensory characteristics, such as tactile sensitivity, were found to correlate with IL-6 (p = 0.027) and IL-1β (p = 0.002) in the ASD group. CONCLUSIONS These findings suggest that CMV-infected children with autism are at increased risk of IL-6 and IL-1β dysregulation, contributing to sensory processing issues and eating behaviours. Further research is needed to enhance CMV testing protocols and better understand the virus's role in the development of sensory and behavioural issues in children with autism.
Collapse
Affiliation(s)
- Isti Anindya
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Rini Sekartini
- Department of Pediatrics, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Ibnu Agus Ariyanto
- Department of Clinical Microbiology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Tjhin Wiguna
- Department of Psychiatry, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Novika Purnama Sari
- Department Clinical & Developmental Neuropsychology, Faculty of Behavioural and Social Sciences, University of Groningen, 9712 TS Groningen, The Netherlands;
| | | | - Amin Soebandrio
- Department of Clinical Microbiology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| |
Collapse
|
131
|
Nataraj BH, Ramesh C, Mallappa RH. Probiotic and postbiotic interference exhibit anti-adhesion effects against clinical methicillin-resistant Staphylococcus aureus (MRSA) and impede MRSA-induced intestinal epithelial hyper-permeability in HT-29 cell line. Microb Pathog 2025; 199:107215. [PMID: 39647539 DOI: 10.1016/j.micpath.2024.107215] [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: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/10/2024]
Abstract
This study investigates the dynamics of MRSA de-colonization on HT-29 cell line using effective strategies like probiotics and postbiotics. Exploring novel alternatives to combat infections caused by antibiotic-resistant pathogens is an urgent need. Harnessing the antagonistic properties of live probiotics and their heat-killed preparations (postbiotics) to curb the growth of AMR pathogens represents a promising and essential area of contemporary research. This study was designed to evaluate the anti-adhesion properties of indigenous probiotics (Limosilactobacillus fermentum Lf1 and Lactiplantibacillus plantarum A5), as well as standard reference strains (Lacticaseibacillus rhamnosus GG and Lactobacillus acidophilus NCFM), and their heat-killed postbiotic preparations against clinical MRSA isolates (MRSA12/206 and 5/255) on the HT-29 cell line. ATR-FTIR-based functional group characterization of the postbiotic preparations revealed the heat-induced alterations in cell surface molecules and architecture. Both probiotic and postbiotic preparations were non-cytotoxic to HT-29 cells. The probiotic intervention, via protective, competitive, and displacement modes, significantly (p < 0.05) reduced the adhesion of MRSA isolates to HT-29 cells, with the protective and competitive modes showing greater efficacy. In contrast, heat-killed probiotics demonstrated notable anti-MRSA adhesion effects across all three modes (protective, competitive, and displacement). In comparison, heat-killed cells exhibited a superior anti-adhesion capability compared to live cells, likely due to the enhanced accessibility of microbe-associated molecular patterns and adhesion sites following heat treatment. Furthermore, co-treatment of MRSA with probiotic strains substantially (p < 0.05) reduced FITC-dextran transflux across the HT-29 cell monolayer. In conclusion, this study highlights the superior anti-adhesion efficacy of heat-killed postbiotics over live probiotic cells against MRSA isolates. It underscores the further need for pre-clinical and in-vivo investigations to validate the anti-MRSA colonization and gut barrier prophylactic or therapeutic potential of the investigated probiotics and postbiotics. Thus, the present study documents and supports the alternative to antibiotics potential of probiotics and postbiotics.
Collapse
Affiliation(s)
- Basavaprabhu Haranahalli Nataraj
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Dairy Bacteriology Section, Southern Regional Station, ICAR-National Dairy Research Institute, Adugodi, 560030, Bengaluru, Karnataka, India.
| | - Chette Ramesh
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Dairy Bacteriology Section, Southern Regional Station, ICAR-National Dairy Research Institute, Adugodi, 560030, Bengaluru, Karnataka, India.
| |
Collapse
|
132
|
de Graav GN, Udomkarnjananun S, Baan CC, Reinders MEJ, Roodnat JI, de Winter BCM, Hesselink DA. New Developments and Therapeutic Drug Monitoring Options in Costimulatory Blockade in Solid Organ Transplantation: A Systematic Critical Review. Ther Drug Monit 2025; 47:64-76. [PMID: 39570574 DOI: 10.1097/ftd.0000000000001275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/31/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE In this review, the authors summarized the latest developments in costimulatory blockade to prevent rejection after solid organ transplantation (SOT) and discussed possibilities for future research and the need for therapeutic drug monitoring (TDM) of these agents. METHODS Studies about costimulatory blockers in SOT in humans or animal transplant models in the past decade (2014-2024) were systematically reviewed in PubMed, European Union clinical trials (EudraCT), and ClinicalTrials.gov . RESULTS Seventy-five registered clinical trials and 58 published articles were found on costimulation blockade of the CD28-CD80/86, CD40-CD40L, and OX40-OX40L pathways. Belatacept, an antagonist of the CD28-CD80/86 pathway, is the only approved costimulatory agent in SOT, hence accounting for most of the research. Other identified costimulatory blocking agents included abatacept and CD28 antagonists tegoprubart, dazodalibep, and TNX-1500. Although tegoprubart was unsuccessful in pancreas transplantation in nonhuman primates, trials in human kidney transplantation are underway. Dazodalibep trials faced recruitment challenges. TNX-1500 was unsuccessful in animal studies and is currently not pursued in humans. After discontinuation of iscalimab (CD40-CD154 pathway antagonist) in SOT, the alternatives, bleselumab and KPL404, showed promising results in kidney transplantation and cardiac xenotransplantation. Studies on secondary costimulatory pathway antagonists, such as OX40-OX40L, have only used animal models. Despite the low interindividual variability in pharmacokinetics (PK) in all studied agents, TDM could be useful for optimizing dosing in PK/pharmacodynamic (PD) studies. CONCLUSIONS The routine use of costimulation blockade in SOT is hindered by problems in efficacy compared with the standard of care. Costimulatory inhibitors could be combined in a calcineurin inhibitor-free regimen. Future PK/pharmacodynamic studies in costimulatory agents and personalized medicine could warrant TDM of these agents.
Collapse
Affiliation(s)
- Gretchen N de Graav
- Department of Internal Medicine, Division of Nephrology, Reinier de Graaf Gasthuis, Delft, the Netherlands
| | - Suwasin Udomkarnjananun
- Department of Medicine, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Carla C Baan
- Transplant Laboratory & Research Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marlies E J Reinders
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| | - Joke I Roodnat
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| |
Collapse
|
133
|
Thiene G. Myocarditis diagnosis: From light microscope to molecular analysis and cardiac magnetic resonance. Eur Heart J Suppl 2025; 27:i61-i66. [PMID: 39980768 PMCID: PMC11836729 DOI: 10.1093/eurheartjsupp/suae106] [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] [Indexed: 02/22/2025]
Abstract
Telling story of myocarditis is characterized by discoveries and inventions. The invention of the microscope opened new avenues in medicine, with the observation of myocardial inflammation by Carl Ludwig Alfred Fiedler. Rudolph Virchow discovered that cells are the elementary units. Karl Albert Ludwig Aschoff first reported rheumatic pancarditis. Gilbert Dallford found enterovirus in the faeces of children, who died suddenly in the village of Coxsackie. Werner Forssmann entered in his own right ventricle with a urologic catheter via the left radial vein. Endomyocardial biopsy, via the femoral or jugular veins, made possible to take away myocardial samples in vivo, for diagnosis of myocarditis or cardiac rejection of transplanted heart. The invention of polymerase chain reaction by Kary Mullis allowed to achieve diagnosis of concealed infections and genetically determined cardiomyopathies. Myocarditis, a significant cause of sudden death, was found to be frequently ascribed to viruses. Cytotoxicity of Coxsackievirus B was proved to consist on released protease 2, encoded by virus genome and cleaving dystrophin. RNA (Coxsackie) and DNA (adenovirus) viruses share a common receptor. Cardiac magnetic resonance revealed to be sensitive and specific in the diagnosis of myocarditis by detecting myocardial oedema. However, it is unable to establish the histotype. The onset of myocarditis may be fulminant; however, extracorporeal membrane oxygenation, invented by Robert Bartlett, allows heart rest, while replacing cardiac contractility. High rates of survival have been achieved, probably because of mild myocardial damage.
Collapse
Affiliation(s)
- Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Via A. Gabelli 61, Padova 35121, Italy
| |
Collapse
|
134
|
Naeem A, Bello MB, Bosaeed M. Insights Into Enterovirus D68 Immunology: Unraveling the Mysteries of Host-Pathogen Interactions. Immun Inflamm Dis 2025; 13:e70117. [PMID: 39912556 PMCID: PMC11800235 DOI: 10.1002/iid3.70117] [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: 06/25/2024] [Revised: 09/03/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Enterovirus D68 (EV-D68) has emerged as a significant respiratory and neurological pathogen, particularly affecting children with severe respiratory illnesses and acute flaccid myelitis. Understanding the interaction between EV-D68 and the host immune system is crucial for developing effective prevention and treatment strategies. OBJECTIVES This review aims to examine the immune response to EV-D68, its mechanisms of immune evasion, and the current progress in vaccine and antiviral development while identifying gaps in knowledge and future research directions. METHODS A comprehensive review of the literature was conducted, focusing on the innate and adaptive immune responses to EV-D68, its strategies for immune evasion, and advancements in therapeutic interventions. RESULTS Pattern recognition receptors detect EV-D68 and trigger antiviral defenses, including interferon production and activation of natural killer cells. B cells generate antibodies, while T cells coordinate a targeted response to the virus. EV-D68 employs mechanisms such as antigenic variation and disruption of host antiviral pathways to evade immune detection. Progress in vaccine and antiviral research shows promise but remains in the early stages. CONCLUSIONS EV-D68 represents a complex and evolving public health challenge. Although the immune system mounts a robust response, the virus's ability to evade these defenses complicates efforts to control it. Continued research is essential to develop effective vaccines and antivirals and to address gaps in understanding its pathogenesis and immune interactions. IMPLICATIONS A multidisciplinary approach is critical to improving diagnostic, preventive, and therapeutic strategies for EV-D68, ensuring better preparedness for future outbreaks.
Collapse
Affiliation(s)
- Asif Naeem
- Infectious Diseases Research DepartmentKing Abdullah International Medical Research CenterRiyadhSaudi Arabia
| | - Muhammad Bashir Bello
- Infectious Diseases Research DepartmentKing Abdullah International Medical Research CenterRiyadhSaudi Arabia
| | - Mohammad Bosaeed
- Infectious Diseases Research DepartmentKing Abdullah International Medical Research CenterRiyadhSaudi Arabia
| |
Collapse
|
135
|
Rubio R, Yavlinsky A, Escalera Zamudio M, Molinos-Albert LM, Martín Pérez C, Pradenas E, Canyelles M, Torres C, Tan C, Swadling L, Ramírez-Morros A, Trinité B, Vidal-Alaball J, Aguilar R, Ruiz-Comellas A, Blanco J, van Dorp L, Balloux F, Dobaño C, Moncunill G. Initial antigen encounter determines robust T-cell immunity against SARS-CoV-2 BA.2.86 variant three years later. J Infect 2025; 90:106402. [PMID: 39746406 DOI: 10.1016/j.jinf.2024.106402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVES We aimed to evaluate the adaptive immune responses cross-recognition of the hypermutated SARS-CoV-2 BA.2.86 variant and identify the determinants influencing this recognition. METHODS We measured BA.2.86 neutralizing antibodies and T-cell responses cross-reactivity in previously exposed participants. We investigated clinical-demographic factors and used a novel in silico analysis to assess viral genetic determinants affecting T-cell responses. RESULTS Despite notable escape from neutralizing antibodies, T-cell responses remained generally preserved, albeit with a significant but small loss in T-cell cross-recognition (7.5%, 14.2%, and 10.8% average loss for IFN-γ, IL-2, and IFN-γ + IL-2, respectively, p<0.05). This is consistent with the prediction of 6 out of 10 immunodominant T-cell epitopes (TCEs) altered by BA.2.86 mutations to have reduced peptide presentation. This effect is expected to be mitigated by total TCEs across the genome. Remarkably, T-cell responses and cross-recognition were 3.5 (IFN-γ), 2 (IL-2) and 2.4 (IFN-γ + IL-2) times higher when first induced by infection rather than by vaccination three years earlier, by increasing number of infections, and by ancestral/Delta than Omicron infections. CONCLUSIONS Our findings underscore the critical role and factors influencing T-cell immunity against evolving SARS-CoV-2 variants, such as first antigen encounter (vaccination or infection), as it is essential for developing effective control strategies.
Collapse
MESH Headings
- Humans
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/immunology
- COVID-19/virology
- Male
- Female
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- T-Lymphocytes/immunology
- Middle Aged
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Cross Reactions/immunology
- Adult
- Aged
- Interleukin-2/immunology
- Antigens, Viral/immunology
- Interferon-gamma/immunology
- Immunity, Cellular
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
Collapse
Affiliation(s)
- Rocío Rubio
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Alexei Yavlinsky
- Institute of Health Informatics, University College London, London, UK
| | | | - Luis M Molinos-Albert
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Carla Martín Pérez
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | | | - Mar Canyelles
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Cèlia Torres
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Cedric Tan
- UCL Genetics Institute, University College London, London, UK
| | - Leo Swadling
- Division of Infection & Immunity, University College London, London, UK
| | - Anna Ramírez-Morros
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain
| | | | - Josep Vidal-Alaball
- Health Promotion in Rural Areas Research Group, Gerència d'Atenció Primària i a la Comunitat Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain; Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada, Gerència d'Atenció Primària i a la Comunitat Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
| | - Ruth Aguilar
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Anna Ruiz-Comellas
- Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, Sant Fruitós de Bages, Spain; Health Promotion in Rural Areas Research Group, Gerència d'Atenció Primària i a la Comunitat Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain; Centre d'Atenció Primària (CAP) Sant Joan de Vilatorrada, Gerència d'Atenció Primària i a la Comunitat Catalunya Central, Institut Català de la Salut, Sant Fruitós de Bages, Spain
| | - Julià Blanco
- IrsiCaixa, Badalona, Spain; Institut Germans Trias I Pujol, IGTPO, Badalona, Spain; Universitat de Vic, Central de Catalunya, UVic-UCC, Vic, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Barcelona, Spain
| | - Lucy van Dorp
- UCL Genetics Institute, University College London, London, UK
| | | | - Carlota Dobaño
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Barcelona, Spain.
| | - Gemma Moncunill
- ISGlobal, Barcelona, Spain; Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Barcelona, Spain.
| |
Collapse
|
136
|
Seguin C, Seif M, Jacoberger-Foissac C, Gentine P, Wantz M, Frisch B, Heurtault B, Fournel S. NOD1 Agonist Induces Proliferation and Plasma Cell Differentiation of Mouse B Cells Especially CD23 high B Cells. Immunol Invest 2025; 54:202-216. [PMID: 39560184 DOI: 10.1080/08820139.2024.2428788] [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: 11/20/2024]
Abstract
BACKGROUND Like innate cells, B cells also express Pattern Recognition Receptors (PRRs) to detect danger signal such as tissue damage or pathogen intrusion. Production of specific antibodies by plasma cells results from the activation and differentiation of B cells following three signals: (i) antigen recognition by B Cell Receptors, (ii) recognition of danger and (iii) T-cell help. However, it is unclear whether T-cell help is dispensable for B cell activation and differentiation or not. Few studies have investigated the role of cytosolic PRRs such as NOD1 in B cell differentiation. METHODS We used splenic C57BL6J B cells to evaluate NOD1 expression and then assessed the effect of stimulation with C12-iE-DAP, a NOD1 ligand, with or without CD40L as a T-cell help signal on B-cell responses globally or according to their CD23 expression level. RESULTS We showed that murine B cells express NOD1 and that the presence of C12-iE-DAP induces activation, proliferation and initiates differentiation in plasma cells even in the absence of a T-dependent signal. Surprisingly, CD23high B cells are more sensitive than CD23low B cells to stimulation. CONCLUSION Our results suggest that the NLR pathway could induce antibody development during infections and be exploited to develop more effective vaccination.
Collapse
Affiliation(s)
- Cendrine Seguin
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Michelle Seif
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Célia Jacoberger-Foissac
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Philippe Gentine
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - May Wantz
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Benoit Frisch
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Béatrice Heurtault
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Sylvie Fournel
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| |
Collapse
|
137
|
Gengiah TN, Heck CJ, Lewis L, Mansoor LE, Harkoo I, Myeni N, Baum MM, Moss JA, Rooney JF, Hankins C, Pozzetto B, Abdool Karim SS, Abdool Karim Q. Acceptability of an annual tenofovir alafenamide implant for HIV prevention in South African women: findings from the CAPRISA 018 Phase I clinical trial. J Int AIDS Soc 2025; 28:e26426. [PMID: 39981604 PMCID: PMC11843155 DOI: 10.1002/jia2.26426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION Long-acting HIV pre-exposure prophylaxis promises to improve uptake, adherence and persistence challenges experienced with daily oral tablets. We assessed the acceptability of an annual tenofovir alafenamide (TAF) implant in South African women enrolled from 9 July 2020 until 31 May 2022 in a Phase I trial. METHODS Six women received one TAF implant for 4 weeks (Group 1), after which 30 women were randomized (4:1, TAF to placebo ratio) to receive 1 or 2 TAF or placebo implants for 48 weeks (Group 2), before trial discontinuation. Acceptability assessments were conducted pre- and post-implant removal. Implant attributes (size, quantity, insertion site, palpability, visibility) and physical experiences (insertion/removal procedures, implant site reactions [ISRs]) were rated on a scale of 1 (highly unacceptable) to 6 (highly acceptable), with 4 being the acceptability threshold. The mean (range) of the mean acceptability scores across all pre-removal visits were calculated, including stratification by removal timing (early vs. scheduled). Implant likes and dislikes were also assessed. RESULTS The median participant age was 26 years. Prior to implant removal, the mean (range) acceptability scores were 5.4 (3.6-6.0) for product attributes and 5.1 (1.7-6.0) for physical experiences. Eleven (31%) participants had early implant removals, occurring on average 19 weeks (range 2-27 weeks) after insertion. The proportion of study visits reporting adherence measure as unacceptable in early versus scheduled removals: ISRs (50% vs. 19%), visibility (30% vs. 15%), palpability (14% vs. 8%), pain (16% vs. 4%) and implant quantity (13% vs. 1%). Pre-removal acceptability scores for ISRs (p = 0.003) and physical experiences (p = 0.05) were significantly associated with early removal. Overall, mean (range) acceptability scores were 5.8 (4.0-6.0) and 5.9 (4.7-6.0) for lifestyle compatibility and likelihood of recommendation, respectively. After removal, 39% of participants found ISRs unacceptable, followed by 22% citing implant visibility. Potential for long-term HIV protection, followed by discreet and convenient use, were most liked, while ISRs were the most disliked aspect. CONCLUSIONS While implant attributes, physical experiences and insertion/removal procedures were largely acceptable, local ISRs significantly reduced tolerability and acceptability, resulting in higher-than-expected early removals. The potential benefits of an annual TAF implant may be undermined unless tolerability is improved.
Collapse
Affiliation(s)
- Tanuja N. Gengiah
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
| | - Craig J. Heck
- Department of EpidemiologyColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
- Division of Infectious Diseases, Department of MedicineColumbia University Irving Medical CenterNew York CityNew YorkUSA
| | - Lara Lewis
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
| | - Leila E. Mansoor
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
| | - Ishana Harkoo
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
| | - Nqobile Myeni
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
| | - Marc M. Baum
- Oak Crest Institute of ScienceMonroviaCaliforniaUSA
| | - John A. Moss
- Oak Crest Institute of ScienceMonroviaCaliforniaUSA
| | | | - Catherine Hankins
- Amsterdam Institute for Global Health and DevelopmentAmsterdamThe Netherlands
| | - Bruno Pozzetto
- Centre International de Recherche en Infectiologie (CIRI), Team GIMAPJean Monnet University of Saint‐EtienneSaint‐EtienneFrance
| | - Salim S. Abdool Karim
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
- Department of EpidemiologyColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South AfricaDurbanSouth Africa
- Department of EpidemiologyColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
| |
Collapse
|
138
|
Kiran NS, Singh S, Yashaswini C, Prajapati BG. Revisiting the potential of natural antimicrobial peptides against emerging respiratory viral disease: a review. 3 Biotech 2025; 15:40. [PMID: 39816617 PMCID: PMC11729606 DOI: 10.1007/s13205-024-04184-3] [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: 07/02/2024] [Accepted: 11/21/2024] [Indexed: 01/18/2025] Open
Abstract
This review assesses the antiviral capabilities of antimicrobial peptides (AMPs) against SARS-CoV-2 and other respiratory viruses, focussing on their therapeutic potential. AMPs, derived from natural sources, exhibit promising antiviral properties by disrupting viral membranes, inhibiting viral entry, and modulating host immune responses. Preclinical studies demonstrate that peptides such as defensins, cathelicidins, and lactoferrin can effectively reduce SARS-CoV-2 replication and inhibit viral spread. In addition, AMPs have shown potential in enhancing the host's antiviral immunity. Despite these promising outcomes, several challenges require assessments before transforming into clinical translation. Several issues related to peptide stability, cytotoxicity, and efficient delivery systems pose significant limitations to their therapeutic application. Recent advancements in peptide engineering, nanotechnology-based delivery systems, and peptide conjugation strategies have improved AMPs stability and bioavailability; however, further optimization is essential. Moreover, whilst AMPs are safe, their effects on host cells and tissues need a thorough investigation to minimise potential adverse reactions. This review concludes that whilst AMPs present a promising route for antiviral therapies, particularly in targeting SARS-CoV-2, extensive clinical trials and additional studies are required to overcome current limitations. Future research should focus on developing more stable, less toxic AMPs formulations with enhanced delivery mechanisms, aiming to integrate AMPs into viable therapeutic options for respiratory viral diseases, including COVID-19 and other emerging infections.
Collapse
Affiliation(s)
| | - Sudarshan Singh
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200 Thailand
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Chandrashekar Yashaswini
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka 560064 India
| | - Bhupendra G. Prajapati
- Shree. S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012 India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000 Thailand
| |
Collapse
|
139
|
Henriot P, El-Kassas M, Anwar W, Girgis SA, El Gaafary M, Jean K, Temime L. An agent-based model to simulate the transmission dynamics of bloodborne pathogens within hospitals. PLoS Comput Biol 2025; 21:e1012850. [PMID: 39993020 PMCID: PMC11882061 DOI: 10.1371/journal.pcbi.1012850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Mathematical models are powerful tools to analyze pathogen spread and assess control strategies in healthcare settings. Nevertheless, available models focus on nosocomial transmission through direct contact or aerosols rather than through blood, even though bloodborne pathogens remain a significant source of iatrogenic infectious risk. Herein, we propose an agent-based SEI (Susceptible-Exposed-Infected) model to reproduce the transmission of bloodborne pathogens dynamically within hospitals. This model simulates the dynamics of patients between hospital wards, from admission to discharge, as well as the dynamics of the devices used during at-risk invasive procedures, considering that patient contamination occurs after exposure to a contaminated device. We first illustrate the use of this model through a case study on hepatitis C virus (HCV) in Egypt. Model parameters, such as HCV upon-admission prevalence and transition probabilities between wards or ward-specific probabilities of undergoing different invasive procedures, are informed with data collected in Ain Shams University Hospital in Cairo. Our results suggest a low risk of HCV acquisition for patients hospitalized in this university hospital. However, we show that in a low-resource hospital, frequent device shortages could lead to increased risk. We also find that systematically screening patients in a few selected high-risk wards could significantly reduce this risk. We then further explore potential model applications through a second illustrative case study based on HBV nosocomial transmission in Ethiopia. In the future, this model could be used to predict the potential burden of emerging bloodborne pathogens and help implement effective control strategies in various hospital contexts.
Collapse
Affiliation(s)
- Paul Henriot
- Laboratoire Modélisation, Épidémiologie Et Surveillance Des Risques Sanitaires, Conservatoire national des arts et métiers (CNAM), Paris, France
- Unité PACRI, Risques Infectieux Et Émergents, CNAM-Institut Pasteur, Paris, France
- UMR EPIA, INRAE, Marcy-l’étoile, France
| | - Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Wagida Anwar
- Department of Community, Environmental and Occupational Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Samia A. Girgis
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha El Gaafary
- Department of Community, Environmental and Occupational Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Kévin Jean
- Laboratoire Modélisation, Épidémiologie Et Surveillance Des Risques Sanitaires, Conservatoire national des arts et métiers (CNAM), Paris, France
- Unité PACRI, Risques Infectieux Et Émergents, CNAM-Institut Pasteur, Paris, France
- IBENS, Ecole normale supérieure, CNRS, INSERM, Université Paris Science & Lettres, Paris, France
| | - Laura Temime
- Laboratoire Modélisation, Épidémiologie Et Surveillance Des Risques Sanitaires, Conservatoire national des arts et métiers (CNAM), Paris, France
- Unité PACRI, Risques Infectieux Et Émergents, CNAM-Institut Pasteur, Paris, France
| |
Collapse
|
140
|
Madlener M, Joost I. [Community acquired bacterial meningitis in adults]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:190-198. [PMID: 39888404 DOI: 10.1007/s00108-025-01851-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
Bacterial meningitis is a rare but severe disease with a high mortality. The most frequent pathogens in adults are pneumococcus, meningococcus and Listeria. The most important key symptoms are headache, meningism and fever; however, the absence of individual cardinal symptoms does not exclude the diagnosis. The empirical treatment consists of ceftriaxone and ampicillin, supplemented with dexamethasone as needed. It should be initiated without delay if bacterial meningitis is suspected. Before this, two pairs of blood cultures should be obtained followed by a lumbar puncture. An elevated intracranial pressure must be excluded via cerebral computed tomography before performing a lumbar puncture only in patients with confirmation of impaired consciousness, focal neurological deficits or epileptic seizures. In such cases treatment is initiated immediately after obtaining blood cultures but before the lumbar puncture. The identification and management of a focus are essential and should be conducted on the day of admission.
Collapse
Affiliation(s)
- Marie Madlener
- Klinik und Poliklinik für Neurologie, Universitätsklinikum Köln, Köln, Deutschland.
| | - Insa Joost
- ABS-Team, Institut für Medizinische Mikrobiologie und Krankenhaushygiene, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| |
Collapse
|
141
|
Han Y, Wang D, Wang Q, Liu Y, Yan M, Ren F, Hu X, Gong R, Li H, He J, Jia Y, Wan J, Long G, Nan K, Huang C, Xu C, Yao Q, Zhang D. Seasonal shifts in respiratory pathogen co-infections and the associated differential induction of cytokines in children. Cytokine 2025; 186:156847. [PMID: 39731898 DOI: 10.1016/j.cyto.2024.156847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
In the post-pandemic era, research on respiratory diseases should refocus on pathogens other than the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Respiratory pathogens, highly infectious to children, with to different modes of infection, such as single-pathogen infections and co-infections. Understanding the seasonal patterns of these pathogens, alongside identifying single infections and co-infections and their impact on the pediatric immune status, is crucial for clinical diagnosis, treatment, and prognosis in children. Our study found that from December 2023 to April 2024, the main co-infection combinations in children shifted from Mycoplasma pneumonia and influenza virus A (MP + IVA) to Bordetella pertussis and rhinovirus (BP + RhV). To explore the impact of these infections, two cohorts were established to analyze the effects of single and co-infections of four respiratory pathogens, MP, IVA, BP, and RhV, on the immune status of pediatric patients. Using multi-cytokine analysis, cytokines, such as PDGF-BB, that were differentially expressed between patients with single and co-infections were identified. Additionally, we observed that children with single-pathogen infections generally exhibited more severe conditions, as evidenced by higher overall cytokine expression than those with co-infections. Our findings provide an important theoretical basis for understanding the pathogenic mechanisms of single and co-infections of respiratory pathogens and clinically differentiating pediatric patients with various respiratory infections.
Collapse
Affiliation(s)
- Yang Han
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Wuhan, Hubei 430023, China
| | - Delong Wang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China; The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, USTC, Hefei, Anhui 230001, China
| | - Qian Wang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Liu
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Mingzhe Yan
- Clinical Laboratory, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Fuli Ren
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Xujuan Hu
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Rui Gong
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huadong Li
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Jingwen He
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Yaling Jia
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Jun Wan
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Gangyu Long
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Kaidi Nan
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Chaolin Huang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China
| | - Congrui Xu
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China.
| | - Qun Yao
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China.
| | - Dingyu Zhang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
142
|
Serban AM, Pepine D, Inceu A, Dadarlat A, Achim A. Embolic risk management in infective endocarditis: predicting the 'embolic roulette'. Open Heart 2025; 12:e003060. [PMID: 39890159 PMCID: PMC11792284 DOI: 10.1136/openhrt-2024-003060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
Life-threatening complications of infective endocarditis (IE,) are heart failure, uncontrolled infection and embolic events (EE), which pose significant morbidity and mortality risks. EE from vegetation rupture are frequent, occurring in more than 50% of patients and can lead to ischaemic stroke and systemic organ infarctions, contributing to poor patient outcomes. Early identification and characterisation of embolic risk factors, including vegetation size, mobility and echogenicity assessed through transthoracic and transoesophageal echocardiography, but also certain pathogens and biomarkers are important for guiding clinical decisions. The latest European Guidelines recommendations emphasise the role of imaging modalities like CT and MRI in detecting silent emboli and guiding therapeutic interventions, including the timely consideration of surgical options to mitigate embolic risks. In this regard, embolic vascular dissemination-including asymptomatic cases detected through multimodality imaging-has been introduced as a new minor criterion for the diagnosis of IE.Depending on the location and severity of the embolism, the embolic risk can either escalate or alternatively, complicate and delay cardiac surgery. The decision to proceed with surgery should not hinge solely on the occurrence of an embolic event, although current guidelines often emphasise this criterion. Therefore, future perspectives should focus on identifying high-risk profiles for EE and investigating whether early surgical intervention benefits these patients, even if they respond favourably to antibiotic therapy. This review explores current literature on echocardiographic and biomarker predictors of EE in IE, aiming to enhance clinical strategies for mitigating embolic complications and improving patient outcomes.
Collapse
Affiliation(s)
- Adela Mihaela Serban
- "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Cardiology Department, Heart Institute Niculae Stăncioiu Cluj-Napoca, Cluj-Napoca, Romania
| | - Diana Pepine
- Cardiology Department, Heart Institute Niculae Stăncioiu Cluj-Napoca, Cluj-Napoca, Romania
| | - Andreea Inceu
- Cardiology Department, Heart Institute Niculae Stăncioiu Cluj-Napoca, Cluj-Napoca, Romania
| | - Alexandra Dadarlat
- "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Cardiology Department, Heart Institute Niculae Stăncioiu Cluj-Napoca, Cluj-Napoca, Romania
| | - Alexandru Achim
- "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Cardiology Department, Heart Institute Niculae Stăncioiu Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
143
|
Aguayo Arelis A, Arana Yepez JE, Rabago Barajas BV, De Los Monteros Conrique FE. Executive functioning in subjects post COVID-19 infection in Mexico. APPLIED NEUROPSYCHOLOGY. ADULT 2025:1-8. [PMID: 39887688 DOI: 10.1080/23279095.2025.2458684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Over the past three years, conflicting evidence has emerged regarding the impact of COVID-19 on executive functions and the frontal lobe. In this study, we evaluated executive functions in individuals from the state of Jalisco who had contracted COVID-19. Sixty individuals with a history of mild COVID-19 were included and compared to historical controls from the Mexican population, who had been assessed prior to the pandemic during the validation of the Trail Making Test Form B, the Stroop Color and Word Test, and the Modified Wisconsin Card Sorting Test (M-WCST). The post-infection group exhibited lower scores only on the M-WCST. Therefore, we concluded that individuals who have recovered from mild COVID-19 do not display widespread impairments in executive functions, with the exception of deficits observed on the M-WCST. This suggests possible neurophysiological alterations in the prefrontal cortex during SARS-CoV-2 infection, given that cognitive flexibility is primarily mediated in this region. These findings contribute to the growing body of evidence indicating that even non-hospitalized COVID-19 patients can experience executive function deficits, providing a foundation for further neurophysiological research into the mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Adriana Aguayo Arelis
- Departamento de Psicologia Aplicada, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico
| | - Jesús Emmanuel Arana Yepez
- Laboratorio de Farmacología y conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Jalisco, Mexico
| | - Brenda Viridiana Rabago Barajas
- Departamento de Psicologia Aplicada, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico
| | | |
Collapse
|
144
|
Canizci Erdemli P, Öcal Demir S, Bozkurt S, Aslan Tuncay S, Yilmaz S, Parlak B, Dizi Işik A, Büyüktaş Aytaç D, Abaci Çapar MÇ, Ergenç Z, Atalay AS, Gökçe İ, Karakoc-Aydiner E, Özen A, Kepenekli E, Akkoç G. Listeria Meningitis as an Indication of Undiagnosed Primary Immune Deficiency, Activated Phosphoinositide 3-Kinase Delta Syndrome: A Case Report. Pediatr Infect Dis J 2025:00006454-990000000-01205. [PMID: 39889727 DOI: 10.1097/inf.0000000000004737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
INTRODUCTION Listeria monocytogenes is a Gram-positive bacillus that causes severe infections mainly in newborns, pregnant women, immunocompromised individuals, and elderly. In this report, we present a case of immune dysregulation that presented with invasive Listeria infection despite the absence of these risk factors. CASE A previously healthy 5-year-old girl developed L. monocytogenes meningitis, which is unusual given her age and lack of typical risk factors. The patient initially presented with fever, diarrhea and altered mental status, unresponsive to empiric antibiotic treatment. Besides clinical diagnosis of meningitis, laboratory tests revealed pleocytosis and positive polymerase chain reaction test for L. monocytogenes in cerebrospinal fluid. Despite initial improvement, the patient developed proteinuria and hypertension and was later diagnosed with focal class 3 lupus nephritis following a renal biopsy. Given the atypical nature of her L. monocytogenes infection, persistent organomegaly, and lupus nephritis, further immunological evaluation was conducted. Genetic testing revealed a de-novo gain-of-function mutation in the PIK3CD gene, confirming the diagnosis of Activated Phosphoinositide 3-Kinase Delta Syndrome 1 (APDS1), a rare primary immunodeficiency characterized by lymphoproliferation and autoimmunity. The patient was started on immunoglobulin replacement therapy and prophylactic trimethoprim-sulfamethoxazole. No recurrence of severe infection occurred during 2 years of follow-up. CONCLUSION This case underscores the importance of considering underlying immune dysregulations in pediatric patients with atypical presentation of Listeria infections.
Collapse
Affiliation(s)
| | | | | | | | - Seyhan Yilmaz
- From the Departments of Pediatric Infectious Diseases
| | - Burcu Parlak
- From the Departments of Pediatric Infectious Diseases
| | | | | | | | - Zeynep Ergenç
- From the Departments of Pediatric Infectious Diseases
| | - Ayşe Sümeyye Atalay
- Pediatric Nephrology, Marmara University School of Medicine, İstanbul, Turkey
| | - İbrahim Gökçe
- Pediatric Nephrology, Marmara University School of Medicine, İstanbul, Turkey
| | | | | | - Eda Kepenekli
- From the Departments of Pediatric Infectious Diseases
| | - Gülşen Akkoç
- From the Departments of Pediatric Infectious Diseases
| |
Collapse
|
145
|
Chemaly RF, Shafat T, Wald A, Kotton CN, Papanicolaou G, Yong MK, Miller V, Komatsu TE, Mullick C, Hodowanec AC, Westman G, Khawaja F, Birkmann A, Ljungman P. Refractory and Resistant Herpes Simplex Virus Mucocutaneous Infections in Immunocompromised Patients: Literature Review and Proposed Definitions for Use in Clinical Trials. Clin Infect Dis 2025:ciae638. [PMID: 39878032 DOI: 10.1093/cid/ciae638] [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: 11/04/2024] [Indexed: 01/31/2025] Open
Abstract
Herpes simplex virus (HSV) infection is one of the most prevalent viral infections worldwide. In general, host immunity is sufficient to clear viral shedding and recurrences, although it is insufficient to prevent subsequent virologic reactivations. In immunocompromised patients, prolonged and difficult-to-treat HSV infections may develop. The diagnosis of refractory HSV infection is based on the lack of clinical response to nucleoside analogs. Antiviral resistance is confirmed via genotypic and/or phenotypic testing. To provide consensus definitions of refractory and/or resistant (R/R) HSV mucocutaneous infections for clinical trial use, the HSV Resistance Working Group of the Transplant Associated Viral Infections Forum, which includes international clinicians, scientists, industry representatives, and regulatory officials, conducted a literature review of previously published data related to R/R HSV infections in immunocompromised patients. We propose definitions of R/R HSV mucocutaneous infections, which will be subject to re-evaluation and revision based on forthcoming data and future studies.
Collapse
Affiliation(s)
- Roy F Chemaly
- Department of Infectious Diseases, Infection Control, and Employee Health, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tali Shafat
- Department of Infectious Diseases, Infection Control, and Employee Health, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anna Wald
- Departments of Medicine, Laboratory Medicine and Pathology, and Epidemiology, University of Washington School of Medicine and Vaccine and Infectious Diseases Division, Fred Cancer Research Center, Seattle, Washington, USA
| | - Camille N Kotton
- Transplant Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Genovefa Papanicolaou
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Michelle K Yong
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Royal Melbourne Hospital, Melbourne, Australia
| | - Veronica Miller
- Forum for Collaborative HIV Research, University of California, Berkeley, California, USA
| | - Takashi E Komatsu
- Division of Antiviral Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Charu Mullick
- Division of Antiviral Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aimee C Hodowanec
- Division of Antiviral Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gabriel Westman
- Swedish Medical Products Agency, Uppsala, Sweden
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Fareed Khawaja
- Department of Infectious Diseases, Infection Control, and Employee Health, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Department of Medicine Huddinge, Division of Hematology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
146
|
Bezerra DT, Mesquita-Ferrari RA, Fernandes KPS, Bussadori SK, Motta LJ, Ando-Suguimoto ES, Frochot C, Perini AM, Rossi F, Mimica MJ, Pereira BJ, Horliana ACRT. Prevalence of Nasal Staphylococcus aureus Carriage in Patients Undergoing Hemodialysis and Assessment of Risk Factors: A Cross-Sectional Study of Outpatients at a University Hospital. Healthcare (Basel) 2025; 13:245. [PMID: 39942434 PMCID: PMC11816970 DOI: 10.3390/healthcare13030245] [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: 11/30/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Infection poses a significant threat of mortality in patients with chronic kidney disease (CKD) undergoing hemodialysis. Staphylococcus aureus (S. aureus) is a common etiological agent, with prior nasal colonization identified as a risk factor for infection. The aim of the present cross-sectional study was to determine the prevalence of nasal S. aureus carriage among patients with CKD undergoing dialysis at a university hospital and identify potential factors associated with colonization. Methods: Nasal swabs were obtained, and bacterial isolates were identified using matrix-assisted laser desorption/ionization time-of-flight spectrometry and antibiogram testing with the Vitek 2 system. Demographic and clinical data were collected for the investigation of risk factors associated with colonization. Results: Among the 96 patients analyzed, 34 were carriers of S. aureus. Among these carriers, three (8.8%) harbored oxacillin-resistant strains. More than half of the S. aureus strains exhibited resistance to clindamycin but susceptibility to oxacillin. Colonization was associated with age and the use of corticosteroids/immunosuppressants. Conclusions: The prevalence of nasal S. aureus carriage was high among patients undergoing dialysis at the university hospital, exceeding that found in the general population. Nasal colonization by S. aureus was linked to corticosteroid use and age. Understanding factors associated with S. aureus nasal colonization in patients on dialysis can assist healthcare providers in preventing the spread of infection and cross-contamination, while reducing risk in this population.
Collapse
Affiliation(s)
- Daniella Teixeira Bezerra
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
| | - Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
- Postgraduate Program in Rehabilitation Sciences, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
| | - Sandra Kalil Bussadori
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
- Postgraduate Program in Rehabilitation Sciences, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil
| | - Lara Jansiski Motta
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
| | - Ellen Sayuri Ando-Suguimoto
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
| | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France;
| | - Alessandra Messina Perini
- Clinical Microbiology Laboratory, Clinical Hospital, University of Sao Paulo Medical School, São Paulo 01246-000, Brazil; (A.M.P.); (F.R.)
| | - Flavia Rossi
- Clinical Microbiology Laboratory, Clinical Hospital, University of Sao Paulo Medical School, São Paulo 01246-000, Brazil; (A.M.P.); (F.R.)
| | - Marcelo Jenne Mimica
- Division of Microbiology, Department of Pathological Sciences, Santa Casa de São Paulo School of Medicine, São Paulo 01224-001, Brazil;
| | - Benedito Jorge Pereira
- Renal Division, Internal Medicine, Clinical Hospital, University of Sao Paulo Medical School, São Paulo 05403-010, Brazil;
| | - Anna Carolina Ratto Tempestini Horliana
- Postgraduate Program in Biophotonics-Medicine, University Nove de Julho, UNINOVE, São Paulo 01504-001, Brazil; (D.T.B.); (R.A.M.-F.); (K.P.S.F.); (S.K.B.); (L.J.M.); (E.S.A.-S.)
| |
Collapse
|
147
|
Meijer EFJ, Marek A, Ramage G, Chowdhary A, Bagrade L, Voss A, Bal AM. A practical approach to investigating nosocomial acquisition of Aspergillus. Med Mycol 2025; 63:myaf007. [PMID: 39875195 DOI: 10.1093/mmy/myaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/12/2025] [Accepted: 01/27/2025] [Indexed: 01/30/2025] Open
Abstract
Invasive mould disease (IMD) has a high mortality in immunosuppressed patients. Invasive aspergillosis (IA) is the most common IMD. A guideline for preventing IA has been published jointly by the Centers for Disease Control and Prevention, the Infectious Disease Society of America, and the American Society of Blood and Marrow Transplantation. Use of high-efficiency particulate air filters, adequate air exchange rates, sealing of patient rooms, and preventing exposure to moulds by nursing patients in areas away from construction sites are recommended by the guideline. However, there is limited information in relation to the actions to be undertaken by infection prevention and control teams in the event of one or more cases of nosocomial aspergillosis. In this review, we describe a systematic approach to aspergillosis by defining possible and probable nosocomial acquisition based on the number of days since hospital admission. We advocate an incremental response to the investigation of nosocomial aspergillosis in patients in protective isolation taking into account the number of cases and the likelihood of nosocomial origin. For single cases of nosocomial IA, we suggest that infection control investigations should focus on case surveillance and walk-through inspection escalating in a stepwise manner to enhanced case surveillance, verification of environmental controls, environmental monitoring, genotyping of clinical and environmental isolates, and review of antifungal prophylaxis for multiple cases and outbreaks. Where applicable, the construction site should be inspected with the aim to reduce the dispersal of conidia. Surveillance systems need to be strengthened to better understand the epidemiology of IA.
Collapse
Affiliation(s)
- Eelco F J Meijer
- Canisius-Wilhelmina Hospital (CWZ)/Dicoon, Medical Microbiology and Immunology, Nijmegen, The Netherlands
- Radboudumc-CWZ Center of Expertise for Mycology, Nijmegen, The Netherlands
- Fungal Infection Working Group, International Society of Antimicrobial Chemotherapy
| | - Aleksandra Marek
- Infection Control Working Group, International Society of Antimicrobial Chemotherapy
- Department of Microbiology, Glasgow Royal Infirmary, Glasgow, UK
- Department of Infection Prevention and Control, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Gordon Ramage
- Safeguarding Health through Infection Prevention (SHIP) Research Group, Research Centre for Health, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Anuradha Chowdhary
- Fungal Infection Working Group, International Society of Antimicrobial Chemotherapy
- Medical Mycology Unit, Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
- National Reference Laboratory for Antimicrobial Resistance in Fungal Pathogens, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Linda Bagrade
- Department of Microbiology, Glasgow Royal Infirmary, Glasgow, UK
- Department of Infection Prevention and Control, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Andreas Voss
- Infection Control Working Group, International Society of Antimicrobial Chemotherapy
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Abhijit M Bal
- Fungal Infection Working Group, International Society of Antimicrobial Chemotherapy
- Department of Infection Prevention and Control, NHS Greater Glasgow and Clyde, Glasgow, UK
- Department of Microbiology, Queen Elizabeth University Hospital, Glasgow, UK
| |
Collapse
|
148
|
Gao J, Pan L, Li P, Liu J, Yang Z, Yang S, Han B, Liu P, Wang C, Chen L, Qu G, Jiang G. Airborne Staphylococcus aureus Exposure Induces Depression-like Behaviors in Mice via Abnormal Neural Oscillation and Mitochondrial Dysfunction. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1133-1144. [PMID: 39772570 DOI: 10.1021/acs.est.4c09497] [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: 01/11/2025]
Abstract
Airborne Staphylococcus aureus exists widely in the natural environment and is closely related to human health. Growing evidence indicates that environmental air pollution elevates the risk of depressive disorders. However, the potential role of airborne S. aureus in the development of depression remains unclear. This study aims to elucidate the neurotoxic effects and potential mechanisms associated with depression caused by airborne S. aureus. Mice were randomly divided into four groups, and the experimental groups with environmental S. aureus were at 4.89 × 102, 8.89 × 105, and 1.27 × 108 CFU/m3 during four consecutive weeks. Airborne S. aureus exposure contributed to depression-like behaviors in mice, especially in the high-concentration group. The electroencephalography signal analysis identified uncoupling of theta and gamma bands and a shift of the beta rhythm toward delta oscillation in the medial prefrontal cortex of mice. Neuropathological analysis showed uplifted neuroinflammation and elevated levels of oxidative stress in the brain. Neuroinflammation and oxidative stress resulted in mitochondrial dysfunction, which could lead to apoptosis. Together, this study provides a strong basis for understanding the adverse outcomes of airborne S. aureus on mental health disorders.
Collapse
Affiliation(s)
- Jie Gao
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Li Pan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Pengxiang Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Jing Liu
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300130, China
| | - Ziye Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Shushuai Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Bin Han
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Ping Liu
- Chongqing Medical University, College of Laboratory Medicine, Chongqing 400016, China
| | - Can Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| |
Collapse
|
149
|
Kholaiq H, Abdelmoumen Y, Moundir A, El Kettani A, Ailal F, Benhsaien I, Adnane F, Drissi Bourhanbour A, Amenzoui N, El Bakkouri J, Bousfiha AA. Human genetic and immunological determinants of SARS-CoV-2 infection and multisystem inflammatory syndrome in children. Clin Exp Immunol 2025; 219:uxae062. [PMID: 39028583 PMCID: PMC11771195 DOI: 10.1093/cei/uxae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/23/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces pneumonia and acute respiratory failure in coronavirus disease 2019 (COVID-19) patients with inborn errors of immunity to type I interferon (IFN-I). The impact of SARS-CoV-2 infection varies widely, ranging from mild respiratory symptoms to life-threatening illness and organ failure, with a higher incidence in men than in women. Approximately 3-5% of critical COVID-19 patients under 60 and a smaller percentage of elderly patients exhibit genetic defects in IFN-I production, including X-chromosome-linked TLR7 and autosomal TLR3 deficiencies. Around 15-20% of cases over 70 years old, and a smaller percentage of younger patients, present with preexisting autoantibodies neutralizing type I interferons. Additionally, innate errors affecting the control of the response to type I interferon have been associated with pediatric multisystem inflammatory syndrome (MIS-C). Several studies have described rare errors of immunity, such as XIAP deficiency, CYBB, SOCS1, OAS1/2, and RNASEL, as underlying factors in MIS-C susceptibility. However, further investigations in expanded patient cohorts are needed to validate these findings and pave the way for new genetic approaches to MIS-C. This review aims to present recent evidence from the scientific literature on genetic and immunological abnormalities predisposing individuals to critical SARS-CoV-2 infection through IFN-I. We will also discuss multisystem inflammatory syndrome in children (MIS-C). Understanding the immunological mechanisms and pathogenesis of severe COVID-19 may inform personalized patient care and population protection strategies against future serious viral infections.
Collapse
Affiliation(s)
- Halima Kholaiq
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Yousra Abdelmoumen
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abderrahmane Moundir
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Assiya El Kettani
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Laboratory of Bacteriology, Virology and Hospital Hygiene, Ibn Rochd University Hospital, Casablanca, Morocco
- Laboratory of Bacteriology and Virology, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Fatima Ailal
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Clinical Immunology and Infectious Pediatrics Department, Abderrahim Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Ibtihal Benhsaien
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Clinical Immunology and Infectious Pediatrics Department, Abderrahim Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Fatima Adnane
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Clinical Immunology and Infectious Pediatrics Department, Abderrahim Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Asmaa Drissi Bourhanbour
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Immunology Laboratory, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Naima Amenzoui
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Clinical Immunology and Infectious Pediatrics Department, Abderrahim Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco
| | - Jalila El Bakkouri
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Immunology Laboratory, Ibn Rochd University Hospital, Casablanca, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Ahmed Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation and Allergies (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Clinical Immunology and Infectious Pediatrics Department, Abderrahim Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco
| |
Collapse
|
150
|
Sokou R, Gounari EA, Tsante KA, Konstantinidi A, Lampridou M, Theodoraki M, Kriebardis AG, Fortis SP, Iacovidou N, Tsantes AG. Thromboelastometry-Based Profiling of Haemostatic Alterations in Neonatal Sepsis by Causative Pathogens. Antibiotics (Basel) 2025; 14:101. [PMID: 39858386 PMCID: PMC11762746 DOI: 10.3390/antibiotics14010101] [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: 11/29/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Neonatal sepsis is a serious condition with high mortality, especially in premature and low-birth-weight neonates. This study aims to examine whether the haemostatic profile of neonates with sepsis defers depending on the type of bacteria (Gram-positive or Gram-negative), by using the method of Rotational Thromboelastometry (ROTEM). Methods: This single-centre prospective cohort study was conducted on 128 neonates with sepsis, including 95 cases caused by Gram-negative pathogens and 33 cases caused by Gram-positive bacteria. All participants were hospitalised in the Neonatal Intensive Care Unit (NICU). ROTEM parameters were compared between neonates with Gram-positive and Gram-negative infections. Results: The ROTEM parameters were found to be significantly different between neonates suffering from Gram-positive versus Gram-negative infections, with Gram-positive pathogens associated with an increased clotting potential compared to Gram-negative pathogens. This is reflected in the higher ROTEM values such as A10, α-angle, and MCF in the EXTEM and INTEM assays. Multivariant analysis showed that Gram-positive infections were linked to increased clot thickness at 10 min (coefficient: 8.9, CI: 2.8-15.0, p = 0.004), higher maximum clot stability (coefficient: 10.4, CI: 4.3-16.6, p = 0.001), and a bigger α-angle (coefficient: 8.0, CI: 2.7-13.2, p = 0.003). Similar findings were observed in the INTEM assay parameters. Conclusions: Neonatal sepsis caused by Gram-positive bacteria leads to a hypercoagulable haemostatic state, whereas neonates with sepsis caused by Gram-negative bacteria exhibit a more hypocoagulable profile and a higher incidence of haemorrhagic episodes. These findings provide valuable insights into the haemostatic disorders associated with sepsis, and may aid in developing an individualised approach for the treatment of those disorders, dependent on and adapted for the specific type of causative organism.
Collapse
Affiliation(s)
- Rozeta Sokou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | | | - Konstantina A. Tsante
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Aikaterini Konstantinidi
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Maria Lampridou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Martha Theodoraki
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 18454 Piraeus, Greece; (A.K.); (M.L.); (M.T.)
| | - Anastasios G. Kriebardis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Sotirios P. Fortis
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece; (K.A.T.); (A.G.K.); (S.P.F.)
| | - Nicoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Andreas G. Tsantes
- Laboratory of Haematology and Blood Bank Unit, “Attikon” Hospital, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece;
- Microbiology Department, “Saint Savvas” Oncology Hospital, 11522 Athens, Greece
| |
Collapse
|