301
|
Pariani E, Piralla A, Pellegrinelli L, Giardina F, Porrello VN, Romano G, Galli C, Sandri L, Ferrari G, Binda S, Vezzosi L, Del Castillo G, Buoro S, Cereda D, Baldanti F. Enhanced laboratory surveillance of respiratory infection disclosed the rapid rise of enterovirus D68 cases, northern Italy, August to September 2024. Euro Surveill 2024; 29:2400645. [PMID: 39392006 PMCID: PMC11484921 DOI: 10.2807/1560-7917.es.2024.29.41.2400645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024] Open
Abstract
We report a considerable increase in enterovirus D68 (EV-D68) cases since July 2024, culminating in an ongoing outbreak of acute respiratory infections in northern Italy, accounting for nearly 90% of all enterovirus infections. The outbreak was identified by community- and hospital-based surveillance systems, detecting EV-D68 in individuals with mild-to-severe respiratory infections. These strains belonged to B3 and a divergent A2 lineage. An increase in adult cases was observed. Enhanced surveillance and molecular characterisation of EV-D68 across Europe are needed.
Collapse
Affiliation(s)
- Elena Pariani
- Department of biomedical sciences for health, University of Milan, Milan, Italy
| | - Antonio Piralla
- Microbiology and virology department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Laura Pellegrinelli
- Department of biomedical sciences for health, University of Milan, Milan, Italy
| | - Federica Giardina
- Department of clinical, surgical, diagnostic and paediatric sciences, University of Pavia, Pavia, Italy
| | - Vincenzo Navobi Porrello
- Department of public health, experimental and forensic medicine, University of Pavia, Pavia, Italy
| | - Greta Romano
- Microbiology and virology department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Cristina Galli
- Department of biomedical sciences for health, University of Milan, Milan, Italy
| | - Laura Sandri
- Department of biomedical sciences for health, University of Milan, Milan, Italy
| | - Guglielmo Ferrari
- Microbiology and virology department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sandro Binda
- Department of biomedical sciences for health, University of Milan, Milan, Italy
| | - Luigi Vezzosi
- Directorate General for Health, Lombardy Region, Milan, Italy
| | | | - Sabrina Buoro
- Directorate General for Health, Lombardy Region, Milan, Italy
| | - Danilo Cereda
- Directorate General for Health, Lombardy Region, Milan, Italy
| | - Fausto Baldanti
- Microbiology and virology department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of clinical, surgical, diagnostic and paediatric sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
302
|
Arango-Franco CA, Ogishi M, Unger S, Delmonte OM, Orrego JC, Yatim A, Velasquez-Lopera MM, Zea-Vera AF, Bohlen J, Chbihi M, Fayand A, Sánchez JP, Rojas J, Seeleuthner Y, Le Voyer T, Philippot Q, Payne KJ, Gervais A, Erazo-Borrás LV, Correa-Londoño LA, Cederholm A, Gallón-Duque A, Goncalves P, Doisne JM, Horev L, Charmeteau-de Muylder B, Álvarez JÁ, Arboleda DM, Pérez-Zapata L, Vásquez-Echeverri E, Moncada-Vélez M, López JA, Caicedo Y, Palterer B, Patiño PJ, Montoya CJ, Chaldebas M, Zhang P, Nguyen T, Ma CS, Jeljeli M, Alzate JF, Cabarcas F, Khan T, Rinchai D, Prétet JL, Boisson B, Marr N, Ibrahim R, Molho-Pessach V, Boisson-Dupuis S, Kiritsi D, Barata JT, Landegren N, Neven B, Abel L, Lisco A, Béziat V, Jouanguy E, Bustamante J, Di Santo JP, Tangye SG, Notarangelo LD, Cheynier R, Natsuga K, Arias AA, Franco JL, Warnatz K, Casanova JL, Puel A. IL-7-dependent and -independent lineages of IL-7R-dependent human T cells. J Clin Invest 2024; 134:e180251. [PMID: 39352394 PMCID: PMC11444196 DOI: 10.1172/jci180251] [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: 02/12/2024] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Infants with biallelic IL7R loss-of-function variants have severe combined immune deficiency (SCID) characterized by the absence of autologous T lymphocytes, but normal counts of circulating B and NK cells (T-B+NK+ SCID). We report 6 adults (aged 22 to 59 years) from 4 kindreds and 3 ancestries (Colombian, Israeli Arab, Japanese) carrying homozygous IL7 loss-of-function variants resulting in combined immunodeficiency (CID). Deep immunophenotyping revealed relatively normal counts and/or proportions of myeloid, B, NK, and innate lymphoid cells. By contrast, the patients had profound T cell lymphopenia, with low proportions of innate-like adaptive mucosal-associated invariant T and invariant NK T cells. They also had low blood counts of T cell receptor (TCR) excision circles, recent thymic emigrant T cells and naive CD4+ T cells, and low overall TCR repertoire diversity, collectively indicating impaired thymic output. The proportions of effector memory CD4+ and CD8+ T cells were high, indicating IL-7-independent homeostatic T cell proliferation in the periphery. Intriguingly, the proportions of other T cell subsets, including TCRγδ+ T cells and some TCRαβ+ T cell subsets (including Th1, Tfh, and Treg) were little affected. Peripheral CD4+ T cells displayed poor proliferation, but normal cytokine production upon stimulation with mitogens in vitro. Thus, inherited IL-7 deficiency impairs T cell development less severely and in a more subset-specific manner than IL-7R deficiency. These findings suggest that another IL-7R-binding cytokine, possibly thymic stromal lymphopoietin, governs an IL-7-independent pathway of human T cell development.
Collapse
Affiliation(s)
- Carlos A Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Susanne Unger
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Julio César Orrego
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Margarita M Velasquez-Lopera
- Sección de Dermatología, Facultad de Medicina, Universidad de Antioquia, Centro de Investigaciones Dermatológicas (CIDERM), Medellín, Antioquia, Colombia
| | - Andrés F Zea-Vera
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
- Clinical Immunology Clinic, Hospital Universitario del Valle, Cali, Colombia
- Microbiology Department, Universidad del Valle, Cali, Colombia
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Marwa Chbihi
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Immunology, Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Fayand
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Juan Pablo Sánchez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - Julian Rojas
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Kathryn J Payne
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Lucia V Erazo-Borrás
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Luis A Correa-Londoño
- Sección de Dermatología, Facultad de Medicina, Universidad de Antioquia, Centro de Investigaciones Dermatológicas (CIDERM), Medellín, Antioquia, Colombia
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alejandro Gallón-Duque
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Pedro Goncalves
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Jean-Marc Doisne
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Liran Horev
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
- Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | | | - Jesús Á Álvarez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Diana M Arboleda
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Lizet Pérez-Zapata
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Estefanía Vásquez-Echeverri
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Marcela Moncada-Vélez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Juan A López
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | | | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Pablo J Patiño
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Carlos J Montoya
- School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Matthieu Chaldebas
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Peng Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Tina Nguyen
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Mohamed Jeljeli
- Cochin University Hospital, Biological Immunology Unit, AP-HP, Paris, France
| | - Juan F Alzate
- Centro Nacional de Secuenciación Genómica CNSG, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Felipe Cabarcas
- Centro Nacional de Secuenciación Genómica CNSG, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Taushif Khan
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jean-Luc Prétet
- Université de Franche-Comté, CNRS, Chrono-environnement & CHU Besançon, Centre National de Référence Papillomavirus, F-25000 Besançon, France
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Ruba Ibrahim
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
| | - Vered Molho-Pessach
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Dimitra Kiritsi
- Department of Dermatology, University Medical Center of Freiburg, Freiburg, Germany
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Bénédicte Neven
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Immunology, Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rémi Cheynier
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Ken Natsuga
- Department of Dermatology, Faculty of Medicine and Graduate of Medicine, Hokkaido University, Sapporo, Japan
| | - Andrés A Arias
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - José Luis Franco
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| |
Collapse
|
303
|
Khan AM, Ehtisham A, Choudhary HH. Cytomegalovirus Colitis Mimicking Ulcerative Colitis Flare in an Immunocompromised Patient: A Challenging Diagnosis. Cureus 2024; 16:e71099. [PMID: 39512970 PMCID: PMC11542870 DOI: 10.7759/cureus.71099] [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: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
This case report details a 40-year-old male patient with a background of ulcerative colitis (UC), who presented with persistent bloody diarrhea refractory to standard treatment. The clinical picture was initially suggestive of a UC flare-up, prompting the continued use of immunosuppressive therapy. However, histopathological evaluation ultimately revealed cytomegalovirus (CMV) colitis, a condition that can mimic UC exacerbation but requires a distinct treatment approach. The patient's immunosuppressive therapy, essential for UC management, likely contributed to the reactivation of latent CMV infection, underscoring the complexity of managing such cases. This case underscores the critical importance of a multidisciplinary approach in differentiating between UC exacerbation and opportunistic infections such as CMV colitis, especially in immunocompromised patients.
Collapse
Affiliation(s)
| | - Amna Ehtisham
- Internal Medicine, Sahiwal Medical College, Sahiwal, PAK
| | | |
Collapse
|
304
|
Huang CY, Su SB, Chen KT. A review of enterovirus-associated hand-foot and mouth disease: preventive strategies and the need for a global enterovirus surveillance network. Pathog Glob Health 2024; 118:538-548. [PMID: 39229797 PMCID: PMC11892072 DOI: 10.1080/20477724.2024.2400424] [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: 09/05/2024] Open
Abstract
Enterovirus (EV)-associated hand, foot, and mouth disease (HFMD) is a significant public health issue worldwide, commonly occurring in children five years of age or younger. The leading causes of most HFMD cases are EVs, which are members of the Picornaviridae family. The typical clinical manifestations of EV-associated HFMD are febrile presentations with mucosal herpangina, oral ulcerations, and skin rashes on the hands and feet. The majority of HFMD cases resolve without consequence; however, a subset progresses to severe neurological and cardiopulmonary complications, which can be fatal. In the past two decades, EV-associated HFMD has received significant attention. In this review, we organize published papers and provide updates on epidemiology, pathogenesis, surveillance, and vaccine developments for EV-associated HFMD. The impact of EV-associated HFMD is increasing globally. Developing efficacious vaccines has become a priority for preventing EV infections without adequate treatment. Simultaneously, emerging EV infections (including EV-D68, EV-A71, Coxsackieviruses, and echoviruses) are increasing, highlighting the need to create a vigilant surveillance system for EV infections worldwide.
Collapse
Affiliation(s)
- Chien-Yuan Huang
- Division of Occupational Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Shih-Bin Su
- Department of Occupational Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Kow-Tong Chen
- Department of Occupational Medicine, Tainan Municipal Hospital (managed by Show Chwan Medical Care Corporation), Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
305
|
Tan X, Gao X, Zheng H, Yuan H, Liu H, Ran Q, Luo M. Platelet dysfunction caused by differentially expressed genes as key pathogenic mechanisms in COVID-19. Minerva Cardiol Angiol 2024; 72:517-534. [PMID: 38804627 DOI: 10.23736/s2724-5683.24.06501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
At the end of 2019, the novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became prevalent worldwide, which brought a heavy medical burden and tremendous economic losses to the world population. In addition to the common clinical respiratory symptoms such as fever, cough and headache, patients with COVID-19 often have hematological diseases, especially platelet dysfunction. Platelet dysfunction usually leads to multiple organ dysfunction, which is closely related to patient severity or mortality. In addition, studies have confirmed significant changes in the gene expression profile of circulating platelets under SARS-CoV-2 infection, which will further lead to changes in platelet function. At the same time, studies have shown that platelets may absorb SARS-COV-2 mRNA independently of ACE2, which further emphasizes the importance of the stability of platelet function in defense against SARS-CoV-2 infection. This study reviewed the relationship between COVID-19 and platelet and SARS-CoV-2 damage to the circulatory system, and further analyzed the significantly differentially expressed mRNA in platelets after infection with SARS-CoV-2 on the basis of previous studies. The top eight hub genes were identified as NLRP3, MT-CO1, CD86, ICAM1, MT-CYB, CASP8, CXCL8 and CXCR4. Subsequently, the effects of SARS-CoV-2 infection on platelet transcript abnormalities and platelet dysfunction were further explored on the basis of 8 hub genes. Finally, the treatment measures of complications caused by platelet dysfunction in patients with COVID-19 were discussed in detail, so as to provide reference for the prevention, diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Xiaoyong Tan
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Huanhuan Zheng
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Hui Yuan
- Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong Liu
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Qijun Ran
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China -
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
306
|
Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, Akanyibah FA, Mao F. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother 2024; 179:117302. [PMID: 39163678 DOI: 10.1016/j.biopha.2024.117302] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex disorder with an unknown cause. However, the dysbiosis of the gut microbiome has been found to play a role in IBD etiology, including exacerbated immune responses and defective intestinal barrier integrity. The gut microbiome can also be a potential biomarker for several diseases, including IBD. Currently, conventional treatments targeting pro-inflammatory cytokines and pathways in IBD-associated dysbiosis do not yield effective results. Other therapies that directly target the dysbiotic microbiome for effective outcomes are emerging. We review the role of the gut microbiome in health and IBD and its potential as a diagnostic, prognostic, and therapeutic target for IBD. This review also explores emerging therapeutic advancements that target gut microbiome-associated alterations in IBD, such as nanoparticle or encapsulation delivery, fecal microbiota transplantation, nutritional therapies, microbiome/probiotic engineering, phage therapy, mesenchymal stem cells (MSCs), gut proteins, and herbal formulas.
Collapse
Affiliation(s)
- Xiu Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu 212300, China
| | - Peipei Cai
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang 212028, China
| | - Anquan Shang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China.
| |
Collapse
|
307
|
Tuncay Tanriverdi S, Gokce EH, Sušanj I, Simić L, Vukelić K, Knežević Z, Ilhan P, Sendemir A, Ozer O. Comprehensive evaluation of xylometazoline hydrochloride formulations: Ex-vivo and in-vitro studies. Eur J Pharm Biopharm 2024; 203:114466. [PMID: 39173937 DOI: 10.1016/j.ejpb.2024.114466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/21/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Xylometazoline is a well-established nasal decongestant that has been used alone and in combination with dexpanthenol as an over the counter (OTC) medicine. Considering the possibility of further improvement of xylometazoline nasal formulations, hyaluronic acid (HA) was evaluated as an additional ingredient. The aim of this study was to investigate the permeation, mucosal retention, and mucoadhesion properties of a new xylometazoline-HA [Xylo-HA] formulation ex vivo and to explore the potential benefits of incorporating HA in the formulation in vitro. Sheep nasal mucosa was used in the ex vivo study, where Xylo-HA was compared with xylometazoline alone [Xylo-Mono], and in combination with dexpanthenol [Xylo-Dex] to understand the impact of formulation changes. The permeation of xylometazoline was generally low (Xylo-Mono 11.14 ± 4.75 %, Xylo-HA 14.57 ± 5.72 % and Xylo-Dex 11.00 ± 3.05 % of the applied dose). The steady state fluxes of xylometazoline were determined as 12.64 ± 3.52 μg/cm2h, 14.94 ± 3.38 μg/cm2h and 12.19 ± 2.05 μg/cm2h for Xylo-Mono, Xylo-HA and Xylo-Dex, respectively. No significant differences were observed between the formulations in the permeation nor mucosal retention studies (p > 0.05 for all), while Xylo-HA exhibited superior mucoadhesive proprieties (p < 0.05 for all). The effects on wound healing and barrier integrity of the three xylometazoline formulations were tested in vitro on HaCaT cells. To better elucidate the role of HA, an additional HA formulation without xylometazoline was prepared (HA-Mono). A scratch test was performed to evaluate wound healing, revealing that the test formulations did not achieve complete wound closure within 72 h and demonstrated a similar effect at the end of the testing period. To assess the effect on barrier integrity, cells were treated for 5 days with daily measurements of transepithelial electrical resistance (TEER). At the end of the experiment, Xylo-Dex showed a moderate 14 % increase in TEER, while Xylo-Mono did not significantly affect this parameter. TEER rose by 951 % in the Xylo-HA, and by 10497 % in the HA group, suggesting that incorporating HA led to enhanced barrier function. Further clinical studies are recommended to better understand the clinical implications and efficacy of the Xylo-HA formulation, with particular focus on the role of HA.
Collapse
Affiliation(s)
- Sakine Tuncay Tanriverdi
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ege University, Bornova, 35100 Izmir, Turkiye.
| | - Evren Homan Gokce
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ege University, Bornova, 35100 Izmir, Turkiye.
| | - Ivan Sušanj
- Jadran Galenski Laboratorij, Svilno 20, Rijeka, Croatia.
| | - Laura Simić
- Jadran Galenski Laboratorij, Svilno 20, Rijeka, Croatia.
| | - Karina Vukelić
- Jadran Galenski Laboratorij, Svilno 20, Rijeka, Croatia.
| | | | - Pelin Ilhan
- Faculty of Engineering, Department of Bioengineering, Ege University, Bornova, 35100 Izmir, Turkiye.
| | - Aylin Sendemir
- Faculty of Engineering, Department of Bioengineering, Ege University, Bornova, 35100 Izmir, Turkiye.
| | - Ozgen Ozer
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Ege University, Bornova, 35100 Izmir, Turkiye.
| |
Collapse
|
308
|
Gervais A, Le Floc'h C, Le Voyer T, Bizien L, Bohlen J, Celmeli F, Al Qureshah F, Masson C, Rosain J, Chbihi M, Lévy R, Castagnoli R, Rothenbuhler A, Jouanguy E, Zhang Q, Zhang SY, Béziat V, Bustamante J, Puel A, Bastard P, Casanova JL. A sensitive assay for measuring whole-blood responses to type I IFNs. Proc Natl Acad Sci U S A 2024; 121:e2402983121. [PMID: 39312669 PMCID: PMC11459193 DOI: 10.1073/pnas.2402983121] [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: 02/12/2024] [Accepted: 08/09/2024] [Indexed: 09/25/2024] Open
Abstract
Human inborn errors of the type I IFN response pathway and auto-Abs neutralizing IFN-α, -β, and/or -ω can underlie severe viral illnesses. We report a simple assay for the detection of both types of condition. We stimulate whole blood from healthy individuals and patients with either inborn errors of type I IFN immunity or auto-Abs against type I IFNs with glycosylated human IFN-α2, -β, or -ω. As controls, we add a monoclonal antibody (mAb) blocking the type I IFN receptors and stimulated blood with IFN-γ (type II IFN). Of the molecules we test, IP-10 (encoded by the interferon-stimulated gene (ISG) CXCL10) is the molecule most strongly induced by type I and type II IFNs in the whole blood of healthy donors in an ELISA-like assay. In patients with inherited IFNAR1, IFNAR2, TYK2, or IRF9 deficiency, IP-10 is induced only by IFN-γ, whereas, in those with auto-Abs neutralizing specific type I IFNs, IP-10 is also induced by the type I IFNs not neutralized by the auto-Abs. The measurement of type I and type II IFN-dependent IP-10 induction therefore constitutes a simple procedure for detecting rare inborn errors of the type I IFN response pathway and more common auto-Abs neutralizing type I IFNs.
Collapse
Affiliation(s)
- Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris, Saint-Louis Hospital, Paris 75010, France
| | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
| | - Fatih Celmeli
- Division of Pediatric Allergy and Immunology, Antalya Education and Research Hospital, University of Medical Science, Antalya 07100, Türkiye
| | - Fahd Al Qureshah
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Cécile Masson
- Bioinformatics Core Facility, Université Paris Cité-Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Marwa Chbihi
- Paris Cité University, Imagine Institute, Paris 75015, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Riccardo Castagnoli
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia 27100, Italy
- Pediatric Clinic, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia 27100, Italy
| | - Anya Rothenbuhler
- Endocrinology and Diabetes for children, Reference Center for rare diseases of calcium and phosphate metabolism, OSCAR network, Platform of expertise for rare diseases of Paris Saclay Hospital, Bicêtre Paris Saclay Hospital, Le Kremlin-Bicêtre 94270, France
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris 75015, France
- Paris Cité University, Imagine Institute, Paris 75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065
- HHMI, New York, NY 10065
- Department of Pediatrics, Necker Hospital for Sick Children, Paris 75015, France
| |
Collapse
|
309
|
Zhang Z, Li X, Huang M, Huang Y, Tan X, Dong Y, Huang Y, Jian J. Siglec7 functions as an inhibitory receptor of non-specific cytotoxic cells and can regulate the innate immune responses in a primitive vertebrate (Oreochromis niloticus). Int J Biol Macromol 2024; 278:134851. [PMID: 39168212 DOI: 10.1016/j.ijbiomac.2024.134851] [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: 05/28/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
In mammals, siglec7, an integral component of the siglecs, is principally found on the surface of natural killer (NK) cells, macrophages, and monocytes, where it interacts with various pathogens to perform immunological regulatory activities. Nonetheless, the immune defense and mechanism of siglec7 in early vertebrates remain unknown. In this study, we identified siglec7 from Oreochromis niloticus (OnSiglec7) and revealed its immune functions. Specifically, OnSiglec7 was abundantly expressed in immune-related tissues of healthy tilapia and its transcription level was strongly activated after being challenged with A. hydrophila, S. agalactiae, and Poly: IC. Meanwhile, OnSiglec7 protein was purified and analyzed, which could recognize multiple pathogens through binding and agglutinating activity. Moreover, OnSiglec7-positive cells were mainly distributed in non-specific cytotoxic cells (NCC) of tilapia HKLs and showed cell membrane localization. Furthermore, OnSiglec7 blockage affected multiple innate immune responses (inflammation, apoptosis, and pyroptosis process) by regulating the activation of MAPK, NF-κB, TLR, and JAK-STAT pathways. Finally, OnSiglec7 blockage also greatly enhanced the cytotoxic effect of tilapia NCC. Summarily, this study uncovers immune functions and mechanisms of siglec7 in primitive vertebrates, thereby enhancing our understanding of the systemic evolution and ancient functions of other siglecs within the host's innate immune system (to our knowledge).
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xing Li
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Meiling Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Xuyan Tan
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yuhang Dong
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
310
|
Sangalli L, Banday F, Sullivan A, Anjum K. Systemic Factors Affecting Prognosis and Outcomes in Periodontal Disease. Dent Clin North Am 2024; 68:571-602. [PMID: 39244245 DOI: 10.1016/j.cden.2024.05.001] [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: 09/09/2024]
Abstract
This review delves into the effects of autoimmune conditions like rheumatoid arthritis, inflammatory disorders such as irritable bowel syndrome, cardiovascular disease, diabetes, infectious ailments like human immunodeficiency virus, and their medications on periodontal therapy outcomes. It also explores the influence of hormones. Understanding these systemic factors is crucial for optimizing periodontal health and treatment efficacy. The review underscores the necessity of considering these variables in periodontal care. Other vital systemic factors are addressed elsewhere in this special edition.
Collapse
Affiliation(s)
- Linda Sangalli
- College of Dental Medicine, Midwestern University, 555 31st, Downers Grove, IL, USA
| | - Fatma Banday
- Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, USA
| | - Andrew Sullivan
- Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, USA
| | - Kainat Anjum
- Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, USA.
| |
Collapse
|
311
|
Zhao K, Wang L, Deng J, Zuo Q, Adila M, Wang X, Dai Z, Tian P. Determining the Disinfectants Resistance Genes and the Susceptibility to Common Disinfectants of Extensively Drug-Resistant Carbapenem-Resistant Klebsiella pneumoniae Strains at a Tertiary Hospital in China. Microb Drug Resist 2024; 30:407-414. [PMID: 39166283 DOI: 10.1089/mdr.2024.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) infection has become a significant threat to global health. The application of chemical disinfectants is an effective infection control strategy to prevent the spread of CRKP in hospital environments. However, bacteria have shown reduced sensitivity to clinical disinfectants in recent years. Furthermore, bacteria can acquire antibiotic resistance due to the induction of disinfectants, posing a considerable challenge to hospital infection prevention and control. This study collected 68 CRKP strains from the Fifth Affiliated Hospital of Xinjiang Medical University in China from 2023 to 2024. These strains were isolated from the sputum, urine, and whole blood samples of patients diagnosed with CRKP infection. Antibiotic susceptibility tests were performed on CRKP strains. Concurrently, the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of disinfectants (benzalkonium bromide, 1% iodophor disinfectant, alcohol, and chlorine-containing disinfectant) against the test isolates were determined by the broth microdilution method. The efflux pump genes (cepA, qacE, qacEΔ1, qacEΔ1-SUL1, oqxA, and oqxB) were detected using polymerase chain reaction. The results showed that 21 out of the 68 CRKP strains exhibited extensive drug resistance, whereas 47 were nonextensively drug-resistant. The MIC value for benzalkonium bromide disinfectants displayed statistically significant differences (p < 0.05) between extensively drug-resistant (XDR) and non-XDR strains. Additionally, the MBC values for benzalkonium bromide disinfectants and 1% iodophor disinfectants displayed statistically significant differences (p < 0.05) between XDR and non-XDR strains. The detection rates for the efflux pump genes were as follows: cepA 52.9%, qacE 39.7%, qacEΔ1 35.2%, qacEΔ1-SUL1 52.9%, oqxA 30.8%, and oqxB 32.3%. The detection rate of the qacEΔ1-SUL1 gene in XDR CRKP strains was significantly higher than in non-XDR CRKP strains (p < 0.05). This indicates a potential link between CRKP bacterial disinfectant efflux pump genes and CRKP bacterial resistance patterns. Ongoing monitoring of the declining sensitivity of XDR strains against disinfectants is essential for the effective control and prevention of superbug.
Collapse
Affiliation(s)
- Kexin Zhao
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Liang Wang
- Department of Laboratory Medicine, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jinglan Deng
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Qiuxia Zuo
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Maimaiti Adila
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Xiao Wang
- School of Nursing, Xinjiang Medical University, Urumqi, China
| | - Zhe Dai
- Fifth School of Clinical Medicine, Xinjiang Medical University, Urumqi, China
| | - Ping Tian
- Infection Management Department, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Health Care Research Center for Xinjiang Regional population, Urumqi, China
| |
Collapse
|
312
|
D'Aleo F, Tuscano A, Servello T, Tripodi M, Abramo C, Bonanno R, Gulino FA, Occhipinti S, Incognito GG, Principe L. Relevance of microbiological cultures of cord blood and placental swabs in the rapid diagnosis of preterm newborn infection due to Listeria monocytogenes: A case report. Case Rep Womens Health 2024; 43:e00638. [PMID: 39188762 PMCID: PMC11345304 DOI: 10.1016/j.crwh.2024.e00638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
Listeria monocytogenes (Lm) is a Gram-positive bacterium causing listeriosis, a rare but severe foodborne infection, particularly impactful during pregnancy. Maternal-fetal transmission can lead to adverse fetal outcomes, yet symptoms in mothers may be nonspecific, delaying intervention. Despite the severity, the mechanisms of vertical transmission remain unclear. This report describes a case of rapid Lm diagnosis in a preterm newborn using cord blood and placental swabs. A 31-week pregnant woman presented with abdominal pain, diarrhea, and reduced fetal movements after consuming raw sushi. Laboratory findings indicated infection, and she vaginally delivered a live infant with placental and fetal abscesses. Cultures confirmed Lm, with swift diagnosis aided by molecular syndromic testing. The neonate received appropriate antibiotics and was asymptomatic by the end of treatment. This case underscores the need for the rapid diagnosis of maternal-fetal listeriosis, as it poses significant risks during pregnancy, including preterm birth and neonatal complications. Current diagnostic methods often delay treatment. This report emphasizes the use of innovative molecular techniques for early diagnosis, which is crucial in managing neonatal infections, especially in preterm newborns.
Collapse
Affiliation(s)
- Francesco D'Aleo
- U.O.C. of Microbiology and Virology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | - Attilio Tuscano
- U.O.C. of Obstetrics and Gynaecology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | - Tarcisio Servello
- U.O.C. of Obstetrics and Gynaecology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | - Marcello Tripodi
- U.O.C. of Obstetrics and Gynaecology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | - Carmela Abramo
- U.O.C. of Obstetrics and Gynaecology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | - Roberta Bonanno
- U.O.C. of Obstetrics and Gynaecology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| | | | - Sara Occhipinti
- Department of General Surgery and Medical Surgical Specialties, University of Catania, Catania, Italy
| | - Giosuè Giordano Incognito
- Department of General Surgery and Medical Surgical Specialties, University of Catania, Catania, Italy
| | - Luigi Principe
- U.O.C. of Microbiology and Virology, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy
| |
Collapse
|
313
|
Wang Q, Wang X, Ding J, Huang L, Wang Z. Structural insight of cell surface sugars in viral infection and human milk glycans as natural antiviral substance. Int J Biol Macromol 2024; 277:133867. [PMID: 39009265 DOI: 10.1016/j.ijbiomac.2024.133867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/24/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Viral infections are caused by the adhesion of viruses to host cell receptors, including sialylated glycans, glycosaminoglycans, and human blood group antigens (HBGAs). Atomic-level structural information on the interactions between viral particles or proteins with glycans can be determined to provide precise targets for designing antiviral drugs. Milk glycans, existing as free oligosaccharides or glycoconjugates, have attracted increasing attention; milk glycans protect infants against infectious diseases, particularly poorly manageable viral infections. Furthermore, several glycans containing structurally distinct sialic acid/fucose/sulfate modifications in human milk acting as a "receptor decoy" and serving as the natural antiviral library, could interrupt virus-receptor interaction in the first line of defense for viral infection. This review highlights the basis of virus-glycan interactions, presents specific glycan receptor binding by gastroenterovirus viruses, including norovirus, enteroviruses, and the breakthroughs in the studies on the antiviral properties of human milk glycans, and also elucidates the role of glycans in respiratory viruses infection. In addition, recent advances in methods for performing virus/viral protein-glycan interactions were reported. Finally, we discuss the prospects and challenges of the studies on the clinical application of human milk glycan for viral interventions.
Collapse
Affiliation(s)
- Qingling Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Jieqiong Ding
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
314
|
Debuysschere C, Nekoua MP, Alidjinou EK, Hober D. The relationship between SARS-CoV-2 infection and type 1 diabetes mellitus. Nat Rev Endocrinol 2024; 20:588-599. [PMID: 38890459 DOI: 10.1038/s41574-024-01004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Environmental factors, in particular viral infections, are thought to have an important role in the pathogenesis of type 1 diabetes mellitus (T1DM). The COVID-19 pandemic reinforced this hypothesis as many observational studies and meta-analyses reported a notable increase in the incidence of T1DM following infection with SARS-CoV-2 as well as an association between SARS-CoV-2 infection and the risk of new-onset T1DM. Experimental evidence suggests that human β-cells express SARS-CoV-2 receptors and that SARS-CoV-2 can infect and replicate in β-cells, resulting in structural or functional alterations of these cells. These alterations include reduced numbers of insulin-secreting granules, impaired pro-insulin (or insulin) secretion, and β-cell transdifferentiation or dedifferentiation. The inflammatory environment induced by local or systemic SARS-CoV-2 infection might result in a set of signals (such as pro-inflammatory cytokines) that lead to β-cell alteration or apoptosis or to a bystander activation of T cells and disruption of peripheral tolerance that triggers autoimmunity. Other mechanisms, such as viral persistence, molecular mimicry and activation of endogenous human retroviruses, are also likely to be involved in the pathogenesis of T1DM following SARS-CoV-2 infection. This Review addresses the issue of the involvement of SARS-CoV-2 infection in the development of T1DM using evidence from epidemiological, clinical and experimental studies.
Collapse
Affiliation(s)
- Cyril Debuysschere
- Université de Lille, CHU Lille, Laboratoire de virologie ULR3610, Lille, France
| | | | | | - Didier Hober
- Université de Lille, CHU Lille, Laboratoire de virologie ULR3610, Lille, France.
| |
Collapse
|
315
|
Eniayewu O, Azuka U, Ogah J, Adejuyigbe E, Bolaji O, Olagunju A. Pharmacogenetics of Efavirenz Exposure in Cervicovaginal Fluid during Pregnancy and Postpartum. Clin Pharmacol Ther 2024; 116:1062-1070. [PMID: 38859656 DOI: 10.1002/cpt.3343] [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: 11/06/2023] [Accepted: 05/22/2024] [Indexed: 06/12/2024]
Abstract
In this study, we investigated the combined influence of pregnancy and genetic polymorphisms on efavirenz pharmacokinetics in cervicovaginal fluid (CVF) of women receiving antiretroviral therapy. Women receiving efavirenz-containing antiretroviral therapy were recruited from two hospitals in Nigeria during 2017-2020. Sparse CVF and plasma samples were obtained during pregnancy to assess the possible association between drug concentration and CYP2B6 polymorphisms (stage I). Participants were stratified into three CYP2B6 516G>T (rs3745274) genotype groups and re-enrolled for intensive pharmacokinetic sampling (stage II). Overall, 159 women (142 pregnant and 12 postpartum) contributed samples in stage I (88 CVF, 81 plasma and 73 paired). CYP2B6 516G>T (rs3745274) remained independently associated with log10 efavirenz CVF concentration during pregnancy after adjusting for plasma concentration, with β (Log10 efavirenz concentration, 95%CI) of 0.204 (0.027, 0.382), P = 0.025). Median (IQR) efavirenz C min in CVF during pregnancy (n = 12) vs. postpartum (n = 12) was 243 ng/mL (168-402) vs. 447 ng/mL (159-974), C max was 1,031 ng/mL (595-1,771) vs. 1,618 ng/mL (675-2,695), and AUC0-24h was 16,465 ng.h/mL (9,356-30,417) vs. 30,715 ng.h/mL (10,980-43,714). CVF-to-plasma AUC ratio was 0.36 during pregnancy and 0.46 postpartum. Upon stratification, efavirenz clearance during pregnancy was 57.9% higher than postpartum in patients with the CYP2B6 516GT genotype; the AUC0-24h and C max were 33.8% and 8.6% lower, respectively. Efavirenz C min in CVF exceeded the protein binding-adjusted IC90 (PBIC90) of 126 ng/mL during pregnancy and postpartum. Efavirenz is well distributed into the CVF; both pregnancy and CYP2B6 polymorphisms affect the extent of exposure.
Collapse
Affiliation(s)
- Oluwasegun Eniayewu
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin, Nigeria
| | - Uche Azuka
- Federal Medical Centre, Makurdi, Nigeria
| | - Jonah Ogah
- Federal Medical Centre, Makurdi, Nigeria
| | - Ebunoluwa Adejuyigbe
- Department of Paediatrics and Child Health, Faculty of Clinical Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Oluseye Bolaji
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Adeniyi Olagunju
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| |
Collapse
|
316
|
Swartling L, Engman ML, Eriksen J, Fischler B, Friman V, Hobell H, Ljungman P, Mellgren K, Navér L, Nyström K, Otto G, Pauksens K, Pettersson K, Rydén I, Westman G, Magnusson J. Management of cytomegalovirus infections - Swedish recommendations 2023. Infect Dis (Lond) 2024; 56:907-917. [PMID: 39185773 DOI: 10.1080/23744235.2024.2392789] [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: 07/18/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024] Open
Abstract
Cytomegalovirus (CMV) infection, which mostly causes a subclinical infection early in life, has important clinical consequences in certain patient groups. CMV is the most common congenital infection and can cause permanent disabilities such as hearing loss and motor- and cognitive deficits in affected infants. In allogeneic haematopoietic stem cell and solid organ transplant recipients, CMV still is an important infectious complication with a risk for life-threatening disease. The previous Swedish recommendations for the management of CMV infections were updated by an expert group under the guidance of The Swedish Reference Group for Antiviral Treatment (RAV) and published at the website of RAV in August 2023 (https://www.sls.se/rav/rekommendationer/cytomegalovirus/). We here provide a translation of the updated recommendations, with minor modifications regarding diagnosis of CMV pneumonia. In the present recommendations, we discuss aspects of old and new CMV antivirals, including dosing for different age groups, and cover the management of congenital infections and CMV in immunocompromised patients. The recommendations are evidence-graded in accordance with the Oxford Centre for Evidence-Based Medicine.
Collapse
Affiliation(s)
- Lisa Swartling
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mona-Lisa Engman
- Department of Pediatrics, Karolinska University Hospital and Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Jaran Eriksen
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Unit of Infectious diseases/Venhälsan, Stockholm South General Hospital, Stockholm, Sweden
| | - Björn Fischler
- Department of Pediatrics, Karolinska University Hospital and Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Vanda Friman
- Department of Infectious Diseases, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hanna Hobell
- Department of Pediatrics, Karolinska University Hospital and Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Karolinska Comprehensive Cancer Center, Stockholm, Sweden
- Division of Hematology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karin Mellgren
- Department of Pediatric Oncology, The Queen Silvia's Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Navér
- Department of Neonatology, Karolinska University Hospital and CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Nyström
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gisela Otto
- Department of Infectious Diseases, Skåne University Hospital, Lund, Sweden
| | - Karlis Pauksens
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Karin Pettersson
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ingehla Rydén
- Department of Pediatrics, Pediatric Drug Therapy Group, Karolinska University Hospital, Stockholm, Sweden
| | - Gabriel Westman
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
- Swedish Medical Products Agency, Uppsala, Sweden
| | - Jesper Magnusson
- Department of Pulmonary Medicine, Institute of Internal Medicine, University of Gothenburg, Gothenburg, Sweden
- The Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
317
|
Bertret C, Maestri F, Salame K, Boutolleau D, Labetoulle M, Rousseau A, Toutée A, Bodaghi B. Acyclovir-resistant bilateral herpetic keratitis in an immunocompromised patient. J Fr Ophtalmol 2024; 47:104283. [PMID: 39294057 DOI: 10.1016/j.jfo.2024.104283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 09/20/2024]
Affiliation(s)
- C Bertret
- Département d'ophtalmologie, hôpital de la Pitié-Salpêtrière, Sorbonne université, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - F Maestri
- Département d'ophtalmologie, hôpital de la Pitié-Salpêtrière, Sorbonne université, 47-83, boulevard de l'Hôpital, 75013 Paris, France.
| | - K Salame
- Département d'ophtalmologie, hôpital de la Pitié-Salpêtrière, Sorbonne université, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - D Boutolleau
- Inserm U1136, service de virologie, centre national de référence Herpèsvirus, hôpital de la Pitié-Salpêtrière, institut Pierre-Louis d'épidémiologie et de santé publique, Sorbonne université, AP-HP, Paris, France
| | - M Labetoulle
- Département d'ophtalmologie, hôpital du Kremlin-Bicêtre, 78, avenue du Général-Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - A Rousseau
- Département d'ophtalmologie, hôpital du Kremlin-Bicêtre, 78, avenue du Général-Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - A Toutée
- Département d'ophtalmologie, hôpital de la Pitié-Salpêtrière, Sorbonne université, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - B Bodaghi
- Département d'ophtalmologie, hôpital de la Pitié-Salpêtrière, Sorbonne université, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| |
Collapse
|
318
|
Tiwari P, Elgazzaz M, Lazartigues E, Hanif K. Effect of Diminazene Aceturate, an ACE2 activator, on platelet CD40L signaling induced glial activation in rat model of hypertension. Int Immunopharmacol 2024; 139:112654. [PMID: 38996777 DOI: 10.1016/j.intimp.2024.112654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.
Collapse
Affiliation(s)
- Priya Tiwari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mona Elgazzaz
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
319
|
Bohoněk M, Máca J, Sagan J, Řezáč D, Fridrich V, Burantová A, Kutáč D, Vabroušek P, Kubů J, Chrdle A, Volfová K, Blahutová Š, Rychlík I, Vonášková K, Majerčin R, Králová R, Štěpánek P, Holub M. Convalescent anti-SARS-CoV-2 plasma for the treatment of patients with COVID-19: a retrospective study RESCOVID-19. Virol J 2024; 21:239. [PMID: 39350163 PMCID: PMC11443855 DOI: 10.1186/s12985-024-02475-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: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Convalescent plasma (CP) collected from people who recovered from COVID-19 became a rapidly available treatment modality in numerous countries, including the Czech Republic. The aims of our study were to evaluate the effectiveness and safety of CP in the treatment of COVID-19. METHODS This retrospective observational study involved six Czech hospitals. This study enrolled patients with and without CP treatment who were hospitalized between April 2020 and April 2021. Propensity score matching and logistic regression analysis were performed to evaluate the influence of CP administration and its timing on the in-hospital survival of COVID-19 patients. RESULTS A total of 1,498 patients were enrolled in the study; 406 (27%) were administered CP, and 1,092 (73%) were not treated with CP. The propensity score-matched control group consisted of 1,218 subjects. The survival of patients treated with CP was 79%, while that of patients in the matched control group was 62% (P<0.001). Moreover, the chance of survival was significantly greater when CP was administered within three days after the onset of COVID-19 symptoms than when CP was administered after four or more days (87% vs. 76%, P <0.001). In addition, adverse effects related to CP administration were recorded in only 2% of patients and were considered mild in all patients. CONCLUSIONS Our study demonstrated that the administration of CP was safe and possibly associated with positive effects that were more pronounced if CP was administered within the first three days after the onset of COVID-19 symptoms.
Collapse
Affiliation(s)
- Miloš Bohoněk
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Jan Máca
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
- Institute of Physiology and Pathological Physiology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jiří Sagan
- Department of Infectious Diseases, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
- Department of Clinical Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - David Řezáč
- The 7th Field Hospital of the Army of the Czech Republic, Hradec Králové, Czech Republic
| | - Viktor Fridrich
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic
| | - Anna Burantová
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic
| | - Dominik Kutáč
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
| | - Pavel Vabroušek
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
| | - Jan Kubů
- Department of Science and Research, University Hospital Bulovka Prague, Prague, Czech Republic
| | - Aleš Chrdle
- Department of Infectious Diseases, České Budejovice Hospital, České Budějovice, Czech Republic
- Faculty of Health and Social Sciences, University of South Bohemia, České Budějovice, Czech Republic
| | - Kateřina Volfová
- Department of Infectious Diseases, České Budejovice Hospital, České Budějovice, Czech Republic
| | - Šárka Blahutová
- Institute of Laboratory Hematology and Transfusion Medicine, Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ivan Rychlík
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Kateřina Vonášková
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Radek Majerčin
- Department of Anesthesiology and Resuscitation, Regional Hospital Jičín, Jičín, Czech Republic
| | - Radka Králová
- Department of Anesthesiology and Resuscitation, Regional Hospital Jičín, Jičín, Czech Republic
| | - Petr Štěpánek
- Department of Anesthesiology and Resuscitation, Regional Hospital Náchod, Náchod, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic.
- Ústřední vojenská nemocnice - Vojenská fakultní nemocnice Praha, U Vojenské nemocnice 1200, Praha 6, 169 06, Czech Republic.
| |
Collapse
|
320
|
Wang N, Yang L, Yuan Y, Wu C, He C. Clinical and Bacterial Characteristics of Bloodstream Infections Caused by Listeria monocytogenes in Western China. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:7785327. [PMID: 39371076 PMCID: PMC11452242 DOI: 10.1155/2024/7785327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/31/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
Objective Bloodstream infections (BSIs) caused by Listeria monocytogenes are linked to high mortality of the patients. Case-specific details related to this disease and causative strains in different districts remain to be characterized. Methods In this study, medical data of BSIs admitted to West China Hospital from October 2017 to March 2023 were retrieved from the hospital information system. The in vitro antimicrobial susceptibility testing and whole-genome sequencing were performed for L. monocytogenes strains isolated from blood specimens. The genetic relationship of these strains with those in public databases was also analyzed. Result The in-hospital mortality of L. monocytogenes BSIs was 25.7% (9/35). The changes in consciousness and elevated serum C-reactive protein (CRP) level were found to be the differential factors of L. monocytogenes BSIs (P < 0.05). All the 27 strains studied were susceptible to ampicillin, meropenem, and erythromycin. Only 22.2% of them were susceptible to trimethoprim-sulfamethoxazole. The Listeria pathogenicity islands 1 (LIPI-1), truncated LIPI-2, and multiple virulence-related genes outside the LIPIs were determined from these strains. Also, 12 sequence types (STs) and 12 clonal complexes (CCs) were identified and classified into clonal lineages I (9/27, 33.3%) and lineages II (18/27, 66.7%), demonstrating genetic differences with the strains in the database. ST451/CC11 (5/27, 18.5%) and ST8/CC8 (4/27, 14.8%) were the common genotypes. Conclusions The consciousness change and elevated serum CRP level were found to be the differential factors of L. monocytogenes BSIs. Considering the high virulence of the strains, it is needed to pay more attention to the dissemination of the predominant genotype.
Collapse
Affiliation(s)
- Nan Wang
- Department of Laboratory MedicineWest China HospitalSichuan University, Chengdu 610041, Sichuan, China
| | - Liuqing Yang
- Department of Laboratory MedicineWest China HospitalSichuan University, Chengdu 610041, Sichuan, China
| | - Yu Yuan
- Department of Laboratory MedicineWest China HospitalSichuan University, Chengdu 610041, Sichuan, China
| | - Chongyang Wu
- Department of Laboratory MedicineWest China HospitalSichuan University, Chengdu 610041, Sichuan, China
| | - Chao He
- Department of Laboratory MedicineWest China HospitalSichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
321
|
Akter D, Biswas J, Miller MJ, Thiele DJ, Murphy EA, O'Connor CM, Moffat JF, Chan GC. Targeting the host transcription factor HSF1 prevents human cytomegalovirus replication in vitro and in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614483. [PMID: 39386472 PMCID: PMC11463536 DOI: 10.1101/2024.09.23.614483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
FDA-approved antivirals against HCMV have several limitations, including only targeting the later stages of the viral replication cycle, adverse side effects, and the emergence of drug-resistant strains. Antivirals targeting host factors specifically activated within infected cells and necessary for viral replication could address the current drawbacks of anti-HCMV standard-of-care drugs. In this study, we found HCMV infection stimulated the activation of the stress response transcription factor heat shock transcription factor 1 (HSF1). HCMV entry into fibroblasts rapidly increased HSF1 activity and subsequent relocalization from the cytoplasm to the nucleus, which was maintained throughout viral replication and in contrast to the transient burst of activity induced by canonical heat shock. Prophylactic pharmacological inhibition or genetic depletion of HSF1 prior to HCMV infection attenuated the expression of all classes of viral genes, including immediate early (IE) genes, and virus production, suggesting HSF1 promotes the earliest stages of the viral replication cycle. Therapeutic treatment with SISU-102, an HSF1 inhibitor tool compound, after IE expression also reduced the levels of L proteins and progeny production, suggesting HSF1 regulates multiple steps along the HCMV replication cycle. Leveraging a newly developed human skin xenograft transplant murine model, we found prophylactic treatment with SISU-102 significantly attenuated viral replication in transplanted human skin xenografts as well as viral dissemination to distal sites. These data demonstrate HCMV infection rapidly activates and relocalizes HSF1 to the nucleus to promote viral replication, which can be exploited as a host-directed antiviral strategy. One Sentence Summary Inhibiting of HSF1 as a host-directed antiviral therapy attenuates HCMV replication in vitro and in vivo.
Collapse
|
322
|
Pagliuca C, Colicchio R, Resta SC, Talà A, Scaglione E, Mantova G, Continisio L, Pagliarulo C, Bucci C, Alifano P, Salvatore P. Neisseria meningitidis activates pyroptotic pathways in a mouse model of meningitis: role of a two-partner secretion system. Front Cell Infect Microbiol 2024; 14:1384072. [PMID: 39376663 PMCID: PMC11456522 DOI: 10.3389/fcimb.2024.1384072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024] Open
Abstract
There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1β and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.
Collapse
Affiliation(s)
- Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Mantova
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Leonardo Continisio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Caterina Pagliarulo
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Pietro Alifano
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- The Institute CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
- Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
323
|
Banse P, Elena SF, Beslon G. Innovation in viruses: fitness valley crossing, neutral landscapes, or just duplications? Virus Evol 2024; 10:veae078. [PMID: 39386076 PMCID: PMC11463231 DOI: 10.1093/ve/veae078] [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: 12/13/2023] [Revised: 07/19/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
Viruses evolve by periods of relative stasis interleaved with sudden, rapid series of mutation fixations, known as evolutionary bursts. These bursts can be triggered by external factors, such as environmental changes, antiviral therapies, or spill-overs from reservoirs into novel host species. However, it has also been suggested that bursts may result from the intrinsic evolutionary dynamics of viruses. Indeed, bursts could be caused by fitness valley crossing, or a neutral exploration of a fitness plateau until an escape mutant is found. In order to investigate the importance of these intrinsic causes of evolutionary bursts, we used a simulation software package to perform massive evolution experiments of viral-like genomes. We tested two conditions: (i) after an external change and (ii) in a constant environment, with the latter condition guaranteeing the absence of an external triggering factor. As expected, an external change was almost systematically followed by an evolutionary burst. However, we also observed bursts in the constant environment as well, albeit much less frequently. We analyzed how many of these bursts are triggered by deleterious, quasi-neutral, or beneficial mutations and show that, while bursts can occasionally be triggered by valley crossing or traveling along neutral ridges, many of them were triggered by chromosomal rearrangements and, in particular, segmental duplications. Our results suggest that combinatorial differences between the different mutation types lead to punctuated evolutionary dynamics, with long periods of stasis occasionally interrupted by short periods of rapid evolution, akin to what is observed in virus evolution.
Collapse
Affiliation(s)
- Paul Banse
- INSA Lyon, INRIA, CNRS, Universite Claude Bernard Lyon 1, Ecole Centrale de Lyon, Université Lumière Lyon 2, LIRIS, UMR5205, Villeurbanne 69621, France
| | - Santiago F Elena
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-Universitat de València, Catedrático Agustín Escardino 9, Paterna, Valencia 46980, Spain
- Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM 87501, USA
| | - Guillaume Beslon
- INSA Lyon, INRIA, CNRS, Universite Claude Bernard Lyon 1, Ecole Centrale de Lyon, Université Lumière Lyon 2, LIRIS, UMR5205, Villeurbanne 69621, France
| |
Collapse
|
324
|
Mokhayeri Y, Taherpour N, Shahbazi F, Ghorbani SS, Fallah S, Etemad K, Izadi N, Mehri A, Farhadi-Babadi K, Rahimi E, Feyzi R, Seifi A, Hashemi Nazari SS. Estimation of outpatient SARS-CoV-2 reinfection and recurrence rates and associated factors among COVID-19 hospitalized patients over one-year old: a multicenter retrospective cohort study. BMC Infect Dis 2024; 24:999. [PMID: 39294562 PMCID: PMC11411993 DOI: 10.1186/s12879-024-09872-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024] Open
Abstract
INTRODUCTION Reinfection with SARS-Cov-2 after recovery can occur that most of them don't require hospitalization. The aim of this study is estimation of out-patient COVID-19 reinfection and recurrence rates and its associated factors among Iranian patients with history of confirmed SARS-Cov-2 infection and hospitalization. METHODS This study is a retrospective cohort conducted from May 2021 to May 2022 in Iran. The national Medical Care Monitoring Center (MCMC) database, obtained from the Ministry of Health and Medical Education, includes all information about confirmed COVID-19 patients who are hospitalized and diagnosed during the pandemic. Using probability proportional to size sampling from 31 provinces, 1,532 patients over one years of age with a history of hospitalization in the MCMC data are randomly selected. After that, interviews by phone are performed with all of the selected patients using a researcher-made questionnaire about the occurrence of overall reinfection without considering the time of infection occurrence, reinfection occurring at least 90 days after the discharge and recurrence (occurring within 90 days after discharge). Univariate and multivariable Cox regression analyses are performed to assess the factors associated with each index. All of the analyses are performed using Stata software version 16. RESULTS In general, 1,532 phone calls are made, out of which 1,095 individuals are willing to participate in the study (response rate ≃ 71%). After assessing the 1,095 patients with a positive history of COVID-19, the rates of non-hospitalized overall SARS-Cov-2 reinfection, reinfection and recurrence are 122.64, 114.09, and 8.55 per 1,000 person-years, respectively. The age range of 19-64 years (aHR:3.93, 95%CI : 1.24-12.41) and COVID-19-related healthcare worker (aHR: 3.67, 95%CI: 1.77-7.61) are identified as risk factors for reinfection, while having comorbidity, being fully vaccinated, and having a partial pressure of oxygen (PaO2) ≥ 93 mmHg during the initial infection are identified as factors that reduce the risk of non-hospitalized reinfection. CONCLUSION Reinfection due to COVID-19 is possible because of the weakened immune system for various reasons and the mutation of the virus. Vaccination, timely boosters, and adherence to preventive measures can help mitigate this risk.
Collapse
Affiliation(s)
- Yaser Mokhayeri
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Niloufar Taherpour
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shahbazi
- Department of Epidemiology, School of Health, Hamadan University of Medical Sciences Hamadan, Hamadan, Iran
| | - Sahar Sotoodeh Ghorbani
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Fallah
- Health Management and Social Development Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Koorosh Etemad
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Izadi
- Research Center for Social Determinants of Health, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Mehri
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kosar Farhadi-Babadi
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Rahimi
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Feyzi
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Seifi
- Department of Infectious Diseases, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Saeed Hashemi Nazari
- Prevention of Cardiovascular Disease Research Center, Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin Ave, Tehran, 198353-5511, Iran.
| |
Collapse
|
325
|
Piechowicz L, Kosznik-Kwaśnicka K, Jarzembowski T, Daca A, Necel A, Bonawenturczak A, Werbowy O, Stasiłojć M, Pałubicka A. Staphylococcus aureus Co-Infection in COVID-19 Patients: Virulence Genes and Their Influence on Respiratory Epithelial Cells in Light of Risk of Severe Secondary Infection. Int J Mol Sci 2024; 25:10050. [PMID: 39337536 PMCID: PMC11431965 DOI: 10.3390/ijms251810050] [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: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pandemics from viral respiratory tract infections in the 20th and early 21st centuries were associated with high mortality, which was not always caused by a primary viral infection. It has been observed that severe course of infection, complications and mortality were often the result of co-infection with other pathogens, especially Staphylococcus aureus. During the COVID-19 pandemic, it was also noticed that patients infected with S. aureus had a significantly higher mortality rate (61.7%) compared to patients infected with SARS-CoV-2 alone. Our previous studies have shown that S. aureus strains isolated from patients with COVID-19 had a different protein profile than the strains in non-COVID-19 patients. Therefore, this study aims to analyze S. aureus strains isolated from COVID-19 patients in terms of their pathogenicity by analyzing their virulence genes, adhesion, cytotoxicity and penetration to the human pulmonary epithelial cell line A549. We have observed that half of the tested S. aureus strains isolated from patients with COVID-19 had a necrotizing effect on the A549 cells. The strains also showed greater variability in terms of their adhesion to the human cells than their non-COVID-19 counterparts.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Tomasz Jarzembowski
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Agnieszka Daca
- Department of Physiopathology, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdansk, Debowa 25, 80-204 Gdansk, Poland
| | - Ada Bonawenturczak
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211 Gdansk, Poland
| | - Anna Pałubicka
- Specialist Hospital in Koscierzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Koscierzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland
| |
Collapse
|
326
|
Eshun G, Osonga FJ, Sadik OA. Quercetin-Derived Platinum Nanomaterials Influence Particle Stability, Catalytic, and Antimicrobial Performance. ACS OMEGA 2024; 9:38557-38568. [PMID: 39310166 PMCID: PMC11411542 DOI: 10.1021/acsomega.4c02948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 09/25/2024]
Abstract
Quercetin possesses high biological properties but low bioavailability, poor solubility, and rapid body clearance. Its structural modification is imperative for enhanced applications. Herein, we demonstrate the catalytic and antimicrobial characteristics of shape-dependent (cuboidal and peanuts) platinum nanoparticles. Modified quercetin, 4'-QP, was employed as the reducing and stabilizing agent for the aqueous synthesis of PtNPs without extraneous reagents. Monodispersed platinum nanocubes (C-PtNPs) and nanopeanuts (P-PtNPs) were produced by reacting 4'-QP and Pt ions in the ratios of 3:1 and 1:1, respectively. TEM characterization confirmed the formation of Pt nanocubes and Pt nanopeanuts, with their corresponding sizes of 39.1 ± 0.20 and 45.1 ± 0.24 nm. The shape-dependency of PtNPs on the nosocomial-causing bacteria, Citrobacter freundii ATCC 8090 (C. freundii) was determined by the Agar well-diffusion assay. Under the same particle size and dose treatments, C-PtNPs and P-PtNPs exhibited 16.28 ± 0.10 and 4.50 ± 0.15 mm zones of inhibition with minimum inhibitory concentrations of 25 and 45 μg/mL, respectively. SEM analysis of C-PtNPs treated C. freundii showed a damaged cell membrane and confirmed contact-killing as the antibacterial mechanism. The catalytic conversion of 4-nitrophenol (4-NP) to 4-amino phenol (4-AP) was tested using a shape-dependent PtNPs catalyst in the presence of sodium borohydride. The conversion rates (k) of C-PtNPs and P-PtNPs in wastewater samples from New Jersey were 0.0108 and 0.00607 s-1, respectively.
Collapse
Affiliation(s)
- Gaddi
B. Eshun
- Department of Chemistry and Environmental
Science BioSMART Center, New Jersey Institute
of Technology, University Heights, 151 Warren Street, Newark, New Jersey 07102, United States
| | - Francis J. Osonga
- Department of Chemistry and Environmental
Science BioSMART Center, New Jersey Institute
of Technology, University Heights, 151 Warren Street, Newark, New Jersey 07102, United States
| | - Omowunmi A. Sadik
- Department of Chemistry and Environmental
Science BioSMART Center, New Jersey Institute
of Technology, University Heights, 151 Warren Street, Newark, New Jersey 07102, United States
| |
Collapse
|
327
|
Chen X, Jin Z, Zhou P, Xie T, Jiang F, Tang Q. Changes of CD8 +CD28 - Tregs and Gamma-Delta-T-cells in a Neonate with Intrauterine Cytomegalovirus Infection: A Case Report. BMC Pediatr 2024; 24:587. [PMID: 39285316 PMCID: PMC11403953 DOI: 10.1186/s12887-024-05051-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Congenital cytomegalovirus (cCMV) infection can lead to a range of adverse outcomes. The majority of cCMV neonates with clinical symptoms are infected postnatally; however, established cases of intrauterine infection are uncommon, resulting in a paucity of reports on clinical findings and lymphocytes expression in CMV-infected neonates. CASE PRESENTATION We followed a neonate with cCMV infection from the onset of hospitalization to several months of follow-up. This infant was intrauterine CMV-positive in the amniotic fluid of the mother at 21 weeks' gestation and received intravenous ganciclovir infusion and sequential oral valganciclovir after birth. The typical clinical signs manifested in the nervous system, liver, and peripheral blood and were documented during the hospitalizaion period and up to the follow-up visit. Flow cytometry was employed to examine the expression of T cells, their subsets, and the associated cytokines in peripheral blood samples at various time points. The flow data for the cCMV neonate were compared with those of the controls at each time point. Following treatment, clinical symptoms improved and the infant became CMV negative. However, developmental delays occurred later in life. The proportion of CD8+CD28- Tregs in the peripheral blood of the neonate with congenital CMV infection was higher than that in the controls at the three time points. The expression levels of perforin and granzyme B secreted by γδ T cells (Vδ1 and Vδ2 T cells), increased during the course of hospitalization until follow-up and were higher than those in the controls at the three time points. CONCLUSIONS Despite the alleviation of clinical symptoms, developmental delay in later life remains inevitable in this intrauterine cCMV neonate. CD8+CD28- Tregs and Vδ1 and Vδ2 T cells secreting perforin and granzyme B may be involved in congenital CMV infection, although this hypothesis requires validation in a larger study. This report may contribute to our understanding of the effect of current treatment and the immune status of intrauterine cCMV-infected neonates.
Collapse
Affiliation(s)
- Xian Chen
- Department of Laboratory Medicine, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Zhenchao Jin
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Ping Zhou
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Tingyan Xie
- Research Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Fan Jiang
- Department of Laboratory Medicine, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China
| | - Quan Tang
- Research Laboratory, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, China.
| |
Collapse
|
328
|
Singh G, Rentsch C, Beattie W, Christensen B, Macrae F, Segal JP. Long-Term Follow Up of Patients Treated for Inflammatory Bowel Disease and Cytomegalovirus Colitis. Diagnostics (Basel) 2024; 14:2030. [PMID: 39335709 PMCID: PMC11431378 DOI: 10.3390/diagnostics14182030] [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: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Pathological reactivation of latent Cytomegalovirus (CMV) is triggered by inflammation and immunosuppression; both present in the pathogenesis and treatment of Inflammatory Bowel Disease (IBD). Whether CMV reactivation is associated with escalating medical therapy, further hospital admissions, or worse clinical outcomes remains controversial. This study aimed to follow up IBD patients with an index episode of CMV colitis and analyse the clinical outcomes. METHODS A retrospective study of patients with IBD treated for CMV colitis was completed. The outcome results were collected at 6-month and 12-month time points after the first episode of CMV colitis. A total of 13 patients with Ulcerative Colitis and 1 with Crohn's Disease were included. RESULTS CMV colitis recurrence occurred in 29% of patients at 12 months. A total of 43% of patients had changed their biologic dose at 6 months and 29% had escalated their biologic dose at 12 months. At 12 months, 36% of patients had been re-hospitalised, including three colectomies. Disease remission was only achieved by 29% of patients at 12 months. CONCLUSIONS IBD patients with CMV colitis have substantial rates of re-hospitalisation, failed medical therapy, and colectomy. These risks may be greater at <6 months from an index episode of CMV colitis.
Collapse
Affiliation(s)
- Gurtej Singh
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
| | - Clarissa Rentsch
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
| | - William Beattie
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
- Department of Gastroenterology, University Hospital Geelong, Geelong 3220, Australia
| | - Britt Christensen
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
- Department of Medicine, The University of Melbourne, Parkville 3010, Australia
| | - Finlay Macrae
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
- Department of Medicine, The University of Melbourne, Parkville 3010, Australia
| | - Jonathan P. Segal
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville 3052, Australia
- Department of Medicine, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
329
|
Sauvat L, Verhoeven PO, Gagnaire J, Berthelot P, Paul S, Botelho-Nevers E, Gagneux-Brunon A. Vaccines and monoclonal antibodies to prevent healthcare-associated bacterial infections. Clin Microbiol Rev 2024; 37:e0016022. [PMID: 39120140 PMCID: PMC11391692 DOI: 10.1128/cmr.00160-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
SUMMARYHealthcare-associated infections (HAIs) represent a burden for public health with a high prevalence and high death rates associated with them. Pathogens with a high potential for antimicrobial resistance, such as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and Clostridioides difficile, are responsible for most HAIs. Despite the implementation of infection prevention and control intervention, globally, HAIs prevalence is stable and they are mainly due to endogenous pathogens. It is undeniable that complementary to infection prevention and control measures, prophylactic approaches by active or passive immunization are needed. Specific groups at-risk (elderly people, chronic condition as immunocompromised) and also healthcare workers are key targets. Medical procedures and specific interventions are known to be at risk of HAIs, in addition to hospital environmental exposure. Vaccines or monoclonal antibodies can be seen as attractive preventive approaches for HAIs. In this review, we present an overview of the vaccines and monoclonal antibodies in clinical development for prevention of the major bacterial HAIs pathogens. Based on the current state of knowledge, we look at the challenges and future perspectives to improve prevention by these means.
Collapse
Affiliation(s)
- Léo Sauvat
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Paul O Verhoeven
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Agents and Hygiene, University-Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julie Gagnaire
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Philippe Berthelot
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Amandine Gagneux-Brunon
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
330
|
Antony F, Kinha D, Nowińska A, Rouse BT, Suryawanshi A. The immunobiology of corneal HSV-1 infection and herpetic stromal keratitis. Clin Microbiol Rev 2024; 37:e0000624. [PMID: 39078136 PMCID: PMC11391706 DOI: 10.1128/cmr.00006-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains.
Collapse
Affiliation(s)
- Ferrin Antony
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Divya Kinha
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Anna Nowińska
- Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
- Ophthalmology Department, Railway Hospital in Katowice, Katowice, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
331
|
Bakowski W, Smiechowicz J, Dragan B, Goździk W, Adamik B. Platelet Aggregation Alterations in Patients with Severe Viral Infection Treated at the Intensive Care Unit: Implications for Mortality Risk. Pathogens 2024; 13:778. [PMID: 39338970 PMCID: PMC11435101 DOI: 10.3390/pathogens13090778] [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: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Severe viral infections often result in abnormal platelet function, affecting various stages of hemostasis. Activated platelets are often considered prothrombotic and more susceptible to further stimulation. However, emerging evidence suggests that initial hyperactivation is followed by platelet exhaustion and hypo-responsiveness, affecting platelet degranulation, activation, and aggregation. We examined early alterations in platelet aggregation among patients (N = 28) with acute respiratory distress syndrome and SARS-CoV-2 infection who were receiving mechanical ventilation and venovenous extracorporeal membrane oxygenation support. Blood samples were stimulated with four different activators: arachidonic acid, adenosine diphosphate, thrombin receptor-activating protein 6, and ristocetin. Our observations revealed that platelet aggregation was reduced in most patients upon admission (ranging from 61 to 89%, depending on the agonist used), and this trend intensified during the 5-day observation period. Concurrently, other coagulation parameters remained within normal ranges, except for elevated d-dimer and fibrinogen levels. Importantly, we found a significant association between platelet aggregation and patient mortality. Impaired platelet aggregation was more severe in patients who ultimately died, and reduced aggregation was associated with a significantly lower probability of survival, as confirmed by Kaplan-Meier analysis (p = 0.028). These findings underscore the potential of aggregometry as an early detection tool for identifying patients at higher risk of mortality within this specific cohort.
Collapse
Affiliation(s)
| | | | | | | | - Barbara Adamik
- Clinical Department of Anesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (W.B.); (J.S.); (B.D.); (W.G.)
| |
Collapse
|
332
|
Bochnia-Bueno L, Coelho GM, Cataneo AHD, Zanluca C, Ferreira LH, Cavalcanti LPDG, Clementino MADF, Yaochite JNU, Dos Santos HG, Nogueira MB, Duarte Dos Santos CN, Raboni SM. Assessment of immune responses to a Comirnaty® booster following CoronaVac® vaccination in healthcare workers. Mem Inst Oswaldo Cruz 2024; 119:e230239. [PMID: 39258622 PMCID: PMC11385826 DOI: 10.1590/0074-02760230239] [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: 12/20/2023] [Accepted: 06/13/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The immunological response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and immunisation is variable. OBJECTIVES To describe the humoral immune response by correlating IgA and IgG antibodies with NAbs titration following CoronaVac® immunisation and an mRNA (Comirnaty®) booster among healthcare workers (HCWs) and to compare the cytokine and interleukin profiles between HCWs vaccinated with CoronaVac and coronavirus disease 2019 (COVID-19) infected patients. METHODS Samples from 133 HCWs collected at 20 (T1) and 90 (T2) days after CoronaVac immunisation and 15 (T3) days after a booster dose with the Comirnaty vaccine were analysed for IgA and IgG EIA and neutralisation assay. Cytokine levels from vaccinated individuals at T1 day and COVID-19 patients were compared. FINDINGS Neutralising antibodies (NAbs) were observed in 81.7% of participants at T1, but only 49.2% maintained detectable NAbs after 90 days. The booster dose increased NAbs response in all participants. The cytokines with the highest levels post-vaccination were IL-6 and MCP-1. The MCP-1, IL-18, and IFN- γ levels were higher in COVID-19 patients than in vaccinated HCWs, while IL-22 levels increased in the vaccinated HCWs group. MAIN CONCLUSIONS The neutralisation titres in the T2 samples decreased, and antibody levels detected at T2 showed a more significant reduction than the neutralisation. The higher IL-22 expression in immunised individuals compared to those with COVID-19 suggests that IL-22 may be beneficial in protecting against severe disease.
Collapse
Affiliation(s)
- Lucas Bochnia-Bueno
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | - Gabriela Mattoso Coelho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | | | - Camila Zanluca
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | - Laura Holtman Ferreira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | | | - Juliana Navarro Ueda Yaochite
- Universidade Federal do Ceará, Faculdade de Farmácia, Odontologia e Enfermagem, Departamento de Análises Clínicas e Toxicologia, Fortaleza, CE, Brasil
| | | | - Meri Bordignon Nogueira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | - Sonia Mara Raboni
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| |
Collapse
|
333
|
Kiss R, Marosi B, Korózs D, Petrik B, Lakatos B, Szabó BG. Clinical and microbiological characteristics and follow-up of invasive Listeria monocytogenes infection among hospitalized patients: real-world experience of 16 years from Hungary. BMC Microbiol 2024; 24:325. [PMID: 39242991 PMCID: PMC11378541 DOI: 10.1186/s12866-024-03478-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
PURPOSE Invasive Listeria monocytogenes infection is rare, but can lead to life-threatening complications among high-risk patients. Our aim was to assess characteristics and follow-up of adults hospitalized with invasive L. monocytogenes infection. METHODS A retrospective observational cohort study was conducted at a national referral center between 2004 and 2019. Patients with proven invasive listeriosis, defined by the European Centre for Disease Prevention and Control criteria, were included. Data collection and follow-up were performed using the hospital electronic system, up until the last documented visit. The primary outcome was in-hospital all-cause mortality, secondary outcomes included residual neurological symptoms, brain abscess occurrence, and requirement for intensive care unit (ICU) admission. RESULTS Altogether, 63 cases were identified (57.1% male, median age 58.8 ± 21.7 years), and 28/63 developed a complicated disease course (44.4%). At diagnosis, 38/63 (60.3%) presented with sepsis, 54/63 (85.7%) had central nervous system involvement, while 9/63 (14.3%) presented with isolated bacteremia. Frequent clinical symptoms included fever (53/63, 84.1%), altered mental state (49/63, 77.8%), with immunocompromised conditions apparent in 56/63 (88.9%). L. monocytogenes was isolated from blood (37/54, 68.5%) and cerebrospinal fluid (48/55, 87.3%), showing in vitro full susceptibility to ampicillin and meropenem (100% each), gentamicin (86.0%) and trimethoprim/sulfamethoxazole (97.7%). In-hospital all-cause mortality was 17/63 (27.0%), and ICU admission was required in 28/63 (44.4%). At discharge, residual neurological deficits (11/46, 23.9%) and brain abscess formation (6/46, 13.0%) were common. CONCLUSION Among hospitalized adult patients with comorbidities, invasive L. monocytogenes infections are associated with high mortality and neurological complications during follow-up.
Collapse
Affiliation(s)
- Rebeka Kiss
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary
| | - Bence Marosi
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary
- Doctoral School of Clinical Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
| | - Dorina Korózs
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary
- Doctoral School of Clinical Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
| | - Borisz Petrik
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary
- Doctoral School of Clinical Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
| | - Botond Lakatos
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary
- Doctoral School of Clinical Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary
- Department of Haematology and Internal Medicine, Division of Infectology, Semmelweis University, Albert Florian ut 5‑7, Budapest, H-1097, Hungary
| | - Bálint Gergely Szabó
- Faculty of Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary.
- South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, H-1097 Albert Florian ut 5-7, Budapest, Hungary.
- Doctoral School of Clinical Medicine, Semmelweis University, H-1085 Ulloi ut 26, Budapest, Hungary.
- Department of Haematology and Internal Medicine, Division of Infectology, Semmelweis University, Albert Florian ut 5‑7, Budapest, H-1097, Hungary.
| |
Collapse
|
334
|
Burford-Gorst CM, Kidd SP. Phenotypic Variation in Staphylococcus aureus during Colonisation Involves Antibiotic-Tolerant Cell Types. Antibiotics (Basel) 2024; 13:845. [PMID: 39335018 PMCID: PMC11428495 DOI: 10.3390/antibiotics13090845] [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: 07/31/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus is a bacterial species that is commonly found colonising healthy individuals but that presents a paradoxical nature: simultaneously, it can migrate within the body and cause a range of diseases. Many of these become chronic by resisting immune responses, antimicrobial treatment, and medical intervention. In part, this ability to persist can be attributed to the adoption of multiple cell types within a single cellular population. These dynamics in the S. aureus cell population could be the result of its interplay with host cells or other co-colonising bacteria-often coagulase-negative Staphylococcal (CoNS) species. Further understanding of the unique traits of S. aureus alternative cell types, the drivers for their selection or formation during disease, as well as their presence even during non-pathological colonisation could advance the development of diagnostic tools and drugs tailored to target specific cells that are eventually responsible for chronic infections.
Collapse
Affiliation(s)
- Chloe M Burford-Gorst
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephen P Kidd
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
335
|
Fernbach S, Mair NK, Abela IA, Groen K, Kuratli R, Lork M, Thorball CW, Bernasconi E, Filippidis P, Leuzinger K, Notter J, Rauch A, Hirsch HH, Huber M, Günthard HF, Fellay J, Kouyos RD, Hale BG. Loss of tolerance precedes triggering and lifelong persistence of pathogenic type I interferon autoantibodies. J Exp Med 2024; 221:e20240365. [PMID: 39017930 PMCID: PMC11253716 DOI: 10.1084/jem.20240365] [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: 02/26/2024] [Revised: 05/10/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
Autoantibodies neutralizing type I interferons (IFN-Is) can underlie infection severity. Here, we trace the development of these autoantibodies at high-resolution using longitudinal samples from 1,876 well-treated individuals living with HIV over a 35-year period. Similar to general populations, ∼1.9% of individuals acquired anti-IFN-I autoantibodies as they aged (median onset ∼63 years). Once detected, anti-IFN-I autoantibodies persisted lifelong, and titers increased over decades. Individuals developed distinct neutralizing and non-neutralizing autoantibody repertoires at discrete times that selectively targeted combinations of IFNα, IFNβ, and IFNω. Emergence of neutralizing anti-IFNα autoantibodies correlated with reduced baseline IFN-stimulated gene levels and was associated with subsequent susceptibility to severe COVID-19 several years later. Retrospective measurements revealed enrichment of pre-existing autoreactivity against other autoantigens in individuals who later developed anti-IFN-I autoantibodies, and there was evidence for prior viral infections or increased IFN at the time of anti-IFN-I autoantibody triggering. These analyses suggest that age-related loss of self-tolerance prior to IFN-I immune-triggering poses a risk of developing lifelong functional IFN-I deficiency.
Collapse
Affiliation(s)
- Sonja Fernbach
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Nina K. Mair
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Irene A. Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kevin Groen
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger Kuratli
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Marie Lork
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Christian W. Thorball
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Ente Ospedaliero Cantonale Lugano, University of Geneva and University of Southern Switzerland, Lugano, Switzerland
| | - Paraskevas Filippidis
- Department of Medicine, Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Julia Notter
- Division of Infectious Diseases, Infection Prevention and Travel Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans H. Hirsch
- Department of Biomedicine, Transplantation and Clinical Virology, University of Basel, Basel, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F. Günthard
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jacques Fellay
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Roger D. Kouyos
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Benjamin G. Hale
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
336
|
Al-Iede M, Ayyad DM, Etoom RA, Aldameiry RH, Toubasi AA. The prevalence and risk factors of methicillin-resistant Staphylococcus aureus among pediatric populations: a systematic review and meta-analysis. Eur J Pediatr 2024; 183:3679-3687. [PMID: 38970703 DOI: 10.1007/s00431-024-05672-7] [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: 03/19/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024]
Abstract
There is scarcity in the data about MRSA prevalence and risk factors among the pediatric population. This research explores the global prevalence and risk factors of MRSA among the pediatric population. PubMed, Scopus, Web of Sciences, and the Cochrane Central Register of Controlled Trials were systematically searched. Our primary goal was to assess the prevalence of MRSA colonization and its related risk factors in the general pediatric population. A total of 124 studies encompassing 44 million participants were included in this meta-analysis, and the overall pooled estimated global prevalence of MRSA colonization in pediatric was 5% [95% CI 4-5%]. Female sex (OR = 4.17; 95% CI, 3.31-5.27), recent surgery (OR = 3.79; 95% CI, 2.20-6.52), recent hospitalization (OR = 2.63; 95% CI, 1.78-3.86), and antibiotic use (OR = 2.42; 95% CI, 1.58-3.72) were significantly associated with higher odds of MRSA colonization. CONCLUSION Future research should build on these findings by emphasizing ongoing efforts to combat MRSA in pediatric settings and implementing targeted interventions. WHAT IS KNOWN • Methicillin-resistant Staphylococcus aureus (MRSA) is considered a threat to public health. It is noteworthy to mention that the prevalence of MRSA strains has not been adequately quantified in many countries, especially in the pediatric population. The pediatric population is a pivotal source of MRSA and may play a central role in its distribution in both community and healthcare settings. A notable study underscores the gravity of the situation, estimating a tenfold increase in the incidence of MRSA infection among children in the USA between 1999 and 2008. WHAT IS NEW • Here we present the first global systematic review and meta-analysis to investigate the prevalence and risk factors of MRSA among the pediatric population. A total of 124 studies encompassing 44 million participants were included in this analysis. The overall pooled estimated global prevalence of MRSA colonization in the pediatric population was 5% [95% CI 4-5%]. The prevalence was the highest in Asia and lowest in Europe. Female sex, recent surgery, recent hospitalization, and antibiotic use were significantly associated with higher odds of MRSA colonization.
Collapse
Affiliation(s)
- Montaha Al-Iede
- School of Medicine, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan.
- Department of Pediatrics, Jordan University Hospital, Amman, Jordan.
- Department of Pediatrics, School of Medicine, The University of Jordan, Amman, Jordan.
| | - Dania M Ayyad
- School of Medicine, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| | - Rasha A Etoom
- School of Medicine, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| | - Rawaby H Aldameiry
- School of Medicine, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| | - Ahmad A Toubasi
- School of Medicine, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| |
Collapse
|
337
|
Reekie J, Stovring H, Nielsen H, Johansen IS, Benfield T, Wiese L, Stærke NB, Iversen K, Mustafa AB, Petersen KT, Juhl MR, Knudsen LS, Iversen MB, Andersen SD, Larsen FD, Baerends EAM, Lindvig SO, Rasmussen LD, Madsen LW, Bannister W, Jensen TO, Dietz LL, Ostrowski SR, Østergaard L, Tolstrup M, Lundgren JD, Søgaard OS. Development of antibody levels and subsequent decline in individuals with vaccine induced and hybrid immunity to SARS-CoV-2. Int J Infect Dis 2024; 146:107111. [PMID: 38801970 DOI: 10.1016/j.ijid.2024.107111] [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: 02/22/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVES This study aimed to compare antibody trajectories among individuals with SARS-CoV-2 hybrid and vaccine-induced immunity. METHODS Danish adults receiving three doses of BTN162b2 or mRNA-1237 were included prior to first vaccination (Day 0). SARS-CoV-2 anti-spike IgG levels were assessed before each vaccine dose, at Day 90, Day 180, 28 days after 3rd vaccination (Day 251), Day 365, and prior to 4th vaccination (Day 535). SARS-CoV-2 PCR results were extracted from the national microbiology database. Mixed-effect multivariable linear regression investigated the impact of hybrid-immunity (stratified into 4 groups: no hybrid immunity, PCR+ prior to 3rd dose, PCR+ after 3rd dose and before Day 365, PCR+ after Day 365) on anti-spike IgG trajectories. RESULTS A total of 4,936 individuals were included, 47% developed hybrid-immunity. Anti-spike IgG increases were observed in all groups at Day 251, with the highest levels in those PCR+ prior to 3rd dose (Geometric Mean; 535,647AU/mL vs. 374,665AU/mL with no hybrid-immunity, P<0.0001). Further increases were observed in participants who developed hybrid immunity after their 3rd dose. Anti-spike IgG levels declined from Day 251-535 in individuals without hybrid-immunity and in those who developed hybrid-immunity prior to their 3rd dose, with lower rate of decline in those with hybrid-immunity. CONCLUSION Hybrid-immunity results in higher and more durable antibody trajectories in vaccinated individuals.
Collapse
Affiliation(s)
- Joanne Reekie
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Henrik Stovring
- Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Isik S Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde, Denmark
| | - Nina Breinholt Stærke
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kasper Iversen
- Department of Cardiology and Department of Emergency Medicine, Herlev, Denmark
| | - Ahmed Basim Mustafa
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Maria Ruwald Juhl
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | | | | | | | - Fredrikke Dam Larsen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Susan Olaf Lindvig
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Lone Wulff Madsen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark; Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Wendy Bannister
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tomas Oestergaard Jensen
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lisa Loksø Dietz
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens D Lundgren
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Schmeltz Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
338
|
Mihalić A, Železnjak J, Lisnić B, Jonjić S, Juranić Lisnić V, Brizić I. Immune surveillance of cytomegalovirus in tissues. Cell Mol Immunol 2024; 21:959-981. [PMID: 39134803 PMCID: PMC11364667 DOI: 10.1038/s41423-024-01186-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 09/01/2024] Open
Abstract
Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.
Collapse
Affiliation(s)
- Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Železnjak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Biomedical Sciences, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
339
|
Bourget-Murray J, Tubin N, Bureau A, Morris J, Ann Azad M, Abdelbary H, Grammatopoulos G, Garceau S. Lower Rates of Reoperation Following Partial or Complete Revision Arthroplasty Compared to Debridement, Antibiotics, and Implant Retention for Early Postoperative and Acute Hematogenous Periprosthetic Hip Infection. J Arthroplasty 2024; 39:2346-2351. [PMID: 38531489 DOI: 10.1016/j.arth.2024.03.054] [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: 07/03/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND This study aimed to: 1) compare treatment outcomes between debridement, antibiotics, and implant retention (DAIR) and partial or complete revision arthroplasty (RA) for early postoperative and acute hematogenous total hip arthroplasty periprosthetic joint infection (PJI) and 2) identify factors associated with treatment outcome. METHODS The study consisted of a retrospective cohort of patients who underwent surgery for PJI between 2004 and 2021. There were 76 patients (74.5%) who underwent DAIR and 26 patients (25.5%) who underwent RA. Treatment success was defined as treatment eradication at a minimum of a 2-year follow up. Bivariate regression analysis was used to assess the effect of different factors on treatment outcomes. Kaplan-Meier survivorship was performed to compare survivorship between cohorts. RESULTS At a mean follow-up of 8.2 years (range, 2.2 to 16.4), significantly more DAIR failed treatment (DAIR, 50 [65.8%]; 10 [38.5%]; P = .015). The 8-year Kaplan-Meier survivorship was 35.1% [95% confidence interval (CI), 24.3 to 45.9] for patients treated with DAIR and 61.5% [95% CI, 42.9 to 80.1] for those treated with RA (log rank = 0.039). Bivariate regression analysis showed performing a RA was associated with a higher likelihood of treatment success (odds ratio 4.499, 95% CI 1.600 to 12.647, P = .004), whereas a higher body mass index was associated with treatment failure (odds ratio 0.934, 95% CI 0.878 to 0.994, P = .032). CONCLUSIONS To reduce the rate of recalcitrant infection following early postoperative or acute hematogenous total hip arthroplasty PJI, RA may be of benefit over DAIR. This is especially relevant in the early postoperative period, when components can be readily exchanged.
Collapse
Affiliation(s)
- Jonathan Bourget-Murray
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Nicholas Tubin
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Antoine Bureau
- Division of Infectious Diseases, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Jared Morris
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Marisa Ann Azad
- Hôtel-Dieu de Lévis, Université Laval, Quebec City, Quebec, Canada
| | - Hesham Abdelbary
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - George Grammatopoulos
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Garceau
- Division of Orthopaedic Surgery, Department of Surgery, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
340
|
Bitu TCV, Silva RC, Bandeira LMP, Cunha DA, Silva PGB, Nobre ÁVV. Stage IV, grade B periodontitis in a patient with coinfection with the human immunodeficiency virus, hepatitis C, and oral candidiasis. J Indian Soc Periodontol 2024; 28:587-590. [PMID: 40134408 PMCID: PMC11932561 DOI: 10.4103/jisp.jisp_301_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/14/2024] [Accepted: 01/24/2025] [Indexed: 03/27/2025] Open
Abstract
Opportunistic infections are common in patients infected with human immunodeficiency virus (HIV) in acquired immunodeficiency syndrome. This case report demonstrates a clinical case of an individual with periodontitis, HIV/hepatitis C coinfection, and candidiasis, using different drugs and under irregular antiretroviral therapy. The combination of several local and systemic conditions highlights the importance of the case and the need for health professionals to know the management of these combined pathologies in the context of HIV infection. We can conclude that it is extremely important for dentists to be aware of periodontal diseases in HIV-infected patients, due to the pathogenic mechanisms associated with these pathologies.
Collapse
|
341
|
Hu JC, Sethi S. New methods to detect bacterial or viral infections in patients with chronic obstructive pulmonary disease. Expert Rev Respir Med 2024; 18:693-707. [PMID: 39175157 PMCID: PMC11583054 DOI: 10.1080/17476348.2024.2396413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Patients with chronic obstructive pulmonary disease (COPD) are frequently colonized and infected by respiratory pathogens. Identifying these infectious etiologies is critical for understanding the microbial dynamics of COPD and for the appropriate use of antimicrobials during exacerbations. AREAS COVERED Traditional methods, such as bacterial and viral cultures, have been standard in diagnosing respiratory infections. However, these methods have significant limitations, including lack of sensitivity and prolonged turnaround time. Modern molecular approaches offer rapid, sensitive, and specific detection, though they also come with their own challenges. This review explores and evaluates the clinical utility of the latest advancements in detecting bacterial and viral respiratory infections in COPD, encompassing molecular techniques, biomarkers, and emerging technologies. EXPERT OPINION In the evolving landscape of COPD management, integrating molecular diagnostics and emerging technologies holds great promise. The enhanced sensitivity of molecular techniques has significantly advanced our understanding of the role of microbes in COPD. However, many of these technologies have primarily been developed for pneumonia diagnosis or research applications, and their clinical utility in managing COPD requires further evaluation.
Collapse
Affiliation(s)
- John C Hu
- Division of Infectious Diseases, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Sanjay Sethi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
342
|
Guo P, Li Z, Cai T, Guo D, Yang B, Zhang C, Shan Z, Wang X, Peng X, Liu G, Shi C, Alharbi M, Alasmari AF. Inhibitory effect and mechanism of oregano essential oil on Listeria monocytogenes cells, toxins and biofilms. Microb Pathog 2024; 194:106801. [PMID: 39025378 DOI: 10.1016/j.micpath.2024.106801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Listeria monocytogenes (L. monocytogenes) is a prevalent foodborne pathogen with a remarkable capacity to form biofilms on utensil surfaces. The Listeriolysin O (LLO) exhibits hemolytic activity, which is responsible for causing human infections. In this study, we investigated the inhibitory effect and mechanism of oregano essential oil (OEO) on L. monocytogenes, evaluated the effects on its biofilm removal and hemolytic activity. The minimum inhibitory concentration (MIC) of OEO against L. monocytogenes was 0.03 % (v/v). L. monocytogenes was treated with OEO at 3/2 MIC for 30 min the bacteria was decreased below the detection limit (10 CFU/mL) in PBS and TSB (the initial bacterial load was about 6.5 log CFU/mL). The level of L. monocytogenes in minced pork co-cultured with OEO (15 MIC) about 2.5 log CFU/g lower than that in the untreated group. The inhibitory mechanisms of OEO against planktonic L. monocytogenes encompassed perturbation of cellular morphology, elevation in reactive oxygen species levels, augmentation of lipid oxidation extent, hyperpolarization of membrane potential, and reduction in intracellular ATP concentration. In addition, OEO reduced biofilm coverage on the surface of glass slides by 62.03 % compared with the untreated group. Meanwhile, OEO (1/8 MIC) treatment reduced the hemolytic activity of L. monocytogenes to 24.6 % compared with the positive control. Molecular docking suggested carvacrol and thymol might reduce the hemolytic activity of L. monocytogenes. The results of this study demonstrate that OEO exhibits inhibitory effects against L. monocytogenes, biofilms and LLO, which had potential as natural antimicrobial for the inhibition of L. monocytogenes.
Collapse
Affiliation(s)
- Peng Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenye Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Ting Cai
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Du Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Baowei Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chunling Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhongguo Shan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Guorong Liu
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, China.
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China; Northwest A&F University ShenZhen Research Institute, Shenzhen, Guangdong, 518057, China.
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
343
|
Peshkova AD, Saliakhutdinova SM, Sounbuli K, Selivanova YA, Andrianova IA, Khabirova AI, Litvinov RI, Weisel JW. The differential formation and composition of leukocyte-platelet aggregates induced by various cellular stimulants. Thromb Res 2024; 241:109092. [PMID: 39024901 PMCID: PMC11411814 DOI: 10.1016/j.thromres.2024.109092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.
Collapse
Affiliation(s)
- Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | | | - Khetam Sounbuli
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Yuliya A Selivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Rustem I Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John W Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
344
|
Liegeon G, Delaugerre C, Molina JM. HIV Pre-Exposure Prophylaxis. Infect Dis Clin North Am 2024; 38:453-474. [PMID: 38871567 DOI: 10.1016/j.idc.2024.04.003] [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: 06/15/2024]
Abstract
Pre-exposure prophylaxis (PrEP) of human immunodeficiency virus (HIV) represents the most significant breakthrough in the HIV prevention field over the past decade. PrEP is an effective strategy in preventing the transmission of HIV across all populations, providing high adherence. The current PrEP options include oral daily and on-demand tenofovir-based regimens, long-acting injections of cabotegravir, and a 1-month dapivirine vaginal ring. As a component of a multifaceted prevention approach, extensive deployment of PrEP holds the promise to significantly reduce the global HIV epidemic. Nonetheless, barriers still exist in terms of uptake, adherence, and persistence, while disparities in PrEP accessibility remain a concern.
Collapse
Affiliation(s)
- Geoffroy Liegeon
- Department of Infectious Diseases and Global Health, University of Chicago Medicine, Office L043 5841 South Maryland Avenue, Chicago 60637, IL, USA.
| | - Constance Delaugerre
- Virology Department, Assistance Publique - Hôpitaux de Paris, Hôpital Saint Louis, Paris, France; Paris Cité University, Paris, France; INSERM UMR 944, Paris, France
| | - Jean-Michel Molina
- Paris Cité University, Paris, France; INSERM UMR 944, Paris, France; Department of Infectious Diseases, Assistance Publique - Hôpitaux de Paris, Hôpitaux Saint Louis et Lariboisière, Paris, France
| |
Collapse
|
345
|
Friedland PL, Tucker S. Phase II Trial of the Impact 0.5% Povidone-Iodine Nasal Spray (Nasodine®) on Shedding of SARS-CoV-2. Laryngoscope 2024; 134:3947-3952. [PMID: 38554057 DOI: 10.1002/lary.31430] [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: 11/29/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/01/2024]
Abstract
OBJECTIVE A Phase II trial was conducted to determine if nasal disinfection with a commercial Good Manufacturing Practice-manufactured 0.5% povidone-iodine nasal spray (Nasodine®) may be a useful adjunct in the management of COVID-19 by reducing viral shedding and prevention of transmission of SARS-CoV-2. The aim was to confirm the results from a human single-dose pilot study by assessing repeated and frequent doses on nasal shedding of SARS-CoV-2 from adult subjects with confirmed COVID-19. METHODS A multicenter, randomized, double-blinded, placebo-controlled Phase II clinical trial involving adults with early COVID-19 symptoms. Baseline nasal swabs were collected to quantify pretreatment SARS-CoV-2 nasal viral load, followed by Nasodine treatment eight times daily over 3 calendar days. Daily nasal swabs were collected post-dose to assess the impact of treatment on nasal viral load, measured by log10 TCID50 in quantitative culture. RESULTS Nasodine subjects exhibited significantly improved reduction in viral load (log10 TCID50) on Days 2-4 compared to placebo recipients (p = 0.028), rate of nasal clearance of viable virus (p = 0.032), and complete (100%) nasal and throat clearance of the virus by Day 5. No difference was seen in antigen shedding as measured by time transition from Rapid Antigen Test (RAT) positivity to RAT negativity. CONCLUSION A total of 20 doses of Nasodine® nasal spray administered over 2.5 days significantly reduced the titers of viable SARS-CoV-2 virus in the nasal passages of COVID-19 subjects. This is the first study demonstrating the efficacy of a tolerable intranasal formulation of povidone-iodine on viral shedding in COVID-19 subjects. Nasal disinfection may diminish viral transmission to others. LEVEL OF EVIDENCE 2 Laryngoscope, 134:3947-3952, 2024.
Collapse
Affiliation(s)
- Peter L Friedland
- Faculty of Medical and Health Sciences, University of Western Australia, Crawley, Western Australia, Australia
- Department Otorhinolaryngology Head Neck Skull Base Surgery, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Firebrick Pharma Limited, Melbourne, Victoria, Australia
| | - Simon Tucker
- Firebrick Pharma Limited, Melbourne, Victoria, Australia
| |
Collapse
|
346
|
Kori M, Kasavi C, Arga KY. Exploring COVID-19 Pandemic Disparities with Transcriptomic Meta-analysis from the Perspective of Personalized Medicine. J Microbiol 2024; 62:785-798. [PMID: 38980578 PMCID: PMC11436439 DOI: 10.1007/s12275-024-00154-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Infection with SARS-CoV2, which is responsible for COVID-19, can lead to differences in disease development, severity and mortality rates depending on gender, age or the presence of certain diseases. Considering that existing studies ignore these differences, this study aims to uncover potential differences attributable to gender, age and source of sampling as well as viral load using bioinformatics and multi-omics approaches. Differential gene expression analyses were used to analyse the phenotypic differences between SARS-CoV-2 patients and controls at the mRNA level. Pathway enrichment analyses were performed at the gene set level to identify the activated pathways corresponding to the differences in the samples. Drug repurposing analysis was performed at the protein level, focusing on host-mediated drug candidates to uncover potential therapeutic differences. Significant differences (i.e. the number of differentially expressed genes and their characteristics) were observed for COVID-19 at the mRNA level depending on the sample source, gender and age of the samples. The results of the pathway enrichment show that SARS-CoV-2 can be combated more effectively in the respiratory tract than in the blood samples. Taking into account the different sample sources and their characteristics, different drug candidates were identified. Evaluating disease prediction, prevention and/or treatment strategies from a personalised perspective is crucial. In this study, we not only evaluated the differences in COVID-19 from a personalised perspective, but also provided valuable data for further experimental and clinical efforts. Our findings could shed light on potential pandemics.
Collapse
Affiliation(s)
- Medi Kori
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752, Istanbul, Turkey.
- Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752, Istanbul, Turkey.
| | - Ceyda Kasavi
- Department of Bioengineering, Marmara University, 34722, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722, Istanbul, Turkey
- Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34722, Istanbul, Turkey
| |
Collapse
|
347
|
Tapela K, Prah DA, Tetteh B, Nuokpem F, Dosoo D, Coker A, Kumi-Ansah F, Amoako E, Assah KO, Kilba C, Nyakoe N, Quansah D, Languon S, Anyigba CA, Ansah F, Agyeman S, Owusu IA, Schneider K, Ampofo WK, Mutungi JK, Amegatcher G, Aniweh Y, Awandare GA, Quashie PK, Bediako Y. Cellular immune response to SARS-CoV-2 and clinical presentation in individuals exposed to endemic malaria. Cell Rep 2024; 43:114533. [PMID: 39052480 PMCID: PMC11372439 DOI: 10.1016/j.celrep.2024.114533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Ghana and other parts of West Africa have experienced lower COVID-19 mortality rates than other regions. This phenomenon has been hypothesized to be associated with previous exposure to infections such as malaria. This study investigated the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the influence of previous malaria exposure. Blood samples were collected from individuals with asymptomatic or symptomatic COVID-19 (n = 217). A variety of assays were used to characterize the SARS-CoV-2-specific immune response, and malaria exposure was quantified using Plasmodium falciparum ELISA. The study found evidence of attenuated immune responses to COVID-19 among asymptomatic individuals, with elevated proportions of non-classical monocytes and greater memory B cell activation. Symptomatic patients displayed higher P. falciparum-specific T cell recall immune responses, whereas asymptomatic individuals demonstrated elevated P. falciparum antibody levels. Summarily, this study suggests that P. falciparum exposure-associated immune modulation may contribute to reduced severity of SARS-CoV-2 infection among people living in malaria-endemic regions.
Collapse
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Diana Ahu Prah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Becky Tetteh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Franklin Nuokpem
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Daniel Dosoo
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Amin Coker
- Accident and Emergency Unit, The Greater Accra Regional Hospital, Accra, Ghana
| | | | - Emmanuella Amoako
- Department of Pediatrics, Cape Coast Teaching Hospital, Cape Coast, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana
| | - Kissi Ohene Assah
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Charlyne Kilba
- Department of Internal Medicine, Surgery, Pediatrics, and Emergency Medicine, Greater Accra Regional Hospital, Accra, Ghana
| | - Nancy Nyakoe
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Darius Quansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana; Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Sylvester Languon
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Claudia Adzo Anyigba
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Felix Ansah
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Seth Agyeman
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana
| | - Irene Amoakoh Owusu
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Kristan Schneider
- Department of Mathematics, Hochschule Mittweida, University of Applied Sciences, Mittweida, Germany
| | - William K Ampofo
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Joe Kimanthi Mutungi
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Gloria Amegatcher
- Department of Medical Laboratory Science, School of Biomedical and Allied Sciences, University of Ghana, Accra, Ghana
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Peter K Quashie
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK.
| | - Yaw Bediako
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana; Yemaachi Biotech Inc., 222 Swaniker St., Accra, Ghana; The Francis Crick Institute, 1 Midland Rd., London NW1 1AT, UK.
| |
Collapse
|
348
|
Chen Y, Zheng Q, Wang H, Tang P, Deng L, Li P, Li H, Hou J, Li J, Wang L, Peng J. Integrating transcriptomics and proteomics to analyze the immune microenvironment of cytomegalovirus associated ulcerative colitis and identify relevant biomarkers. BioData Min 2024; 17:26. [PMID: 39192288 DOI: 10.1186/s13040-024-00382-0] [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: 07/15/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND In recent years, significant morbidity and mortality in patients with severe inflammatory bowel disease (IBD) and cytomegalovirus (CMV) have drawn considerable attention to the status of CMV infection in the intestinal mucosa of IBD patients and its role in disease progression. However, there is currently no high-throughput sequencing data for ulcerative colitis patients with CMV infection (CMV + UC), and the immune microenvironment in CMV + UC patients have yet to be explored. METHOD The xCell algorithm was used for evaluate the immune microenvironment of CMV + UC patients. Then, WGCNA analysis was explored to obtain the co-expression modules between abnormal immune cells and gene level or protein level. Next, three machine learning approach include Random Forest, SVM-rfe, and Lasso were used to filter candidate biomarkers. Finally, Best Subset Selection algorithms was performed to construct the diagnostic model. RESULTS In this study, we performed transcriptomic and proteomic sequencing on CMV + UC patients to establish a comprehensive immune microenvironment profile and found 11 specific abnormal immune cells in CMV + UC group. After using multi-omics integration algorithms, we identified seven co-expression gene modules and five co-expression protein modules. Subsequently, we utilized various machine learning algorithms to identify key biomarkers with diagnostic efficacy and constructed an early diagnostic model. We identified a total of eight biomarkers (PPP1R12B, CIRBP, CSNK2A2, DNAJB11, PIK3R4, RRBP1, STX5, TMEM214) that play crucial roles in the immune microenvironment of CMV + UC and exhibit superior diagnostic performance for CMV + UC. CONCLUSION This 8 biomarkers model offers a new paradigm for the diagnosis and treatment of IBD patients post-CMV infection. Further research into this model will be significant for understanding the changes in the host immune microenvironment following CMV infection.
Collapse
Affiliation(s)
- Yang Chen
- Yunnan Provincial Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Qingqing Zheng
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Hui Wang
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Peiren Tang
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Li Deng
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Pu Li
- Department of General Practice, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
| | - Huan Li
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Jianhong Hou
- Department of Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China.
| | - Jie Li
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Li Wang
- Department of Pathology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China.
- Department of Pathology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China.
| | - Jun Peng
- Department of Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China.
| |
Collapse
|
349
|
Zheng H, Wu S, Chen W, Cai S, Zhan M, Chen C, Lin J, Xie Z, Ou J, Ye W. Meta-analysis of hybrid immunity to mitigate the risk of Omicron variant reinfection. Front Public Health 2024; 12:1457266. [PMID: 39253287 PMCID: PMC11381385 DOI: 10.3389/fpubh.2024.1457266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Background Hybrid immunity (a combination of natural and vaccine-induced immunity) provides additional immune protection against the coronavirus disease 2019 (COVID-19) reinfection. Today, people are commonly infected and vaccinated; hence, hybrid immunity is the norm. However, the mitigation of the risk of Omicron variant reinfection by hybrid immunity and the durability of its protection remain uncertain. This meta-analysis aims to explore hybrid immunity to mitigate the risk of Omicron variant reinfection and its protective durability to provide a new evidence-based basis for the development and optimization of immunization strategies and improve the public's awareness and participation in COVID-19 vaccination, especially in vulnerable and at-risk populations. Methods Embase, PubMed, Web of Science, Chinese National Knowledge Infrastructure, and Wanfang databases were searched for publicly available literature up to 10 June 2024. Two researchers independently completed the data extraction and risk of bias assessment and cross-checked each other. The Newcastle-Ottawa Scale assessed the risk of bias in included cohort and case-control studies, while criteria recommended by the Agency for Health Care Research and Quality (AHRQ) evaluated cross-sectional studies. The extracted data were synthesized in an Excel spreadsheet according to the predefined items to be collected. The outcome was Omicron variant reinfection, reported as an Odds Ratio (OR) with its 95% confidence interval (CI) and Protective Effectiveness (PE) with 95% CI. The data were pooled using a random- or fixed-effects model based on the I2 test. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Results Thirty-three articles were included. Compared with the natural immunity group, the hybrid immunity (booster vaccination) group had the highest level of mitigation in the risk of reinfection (OR = 0.43, 95% CI:0.34-0.56), followed by the complete vaccination group (OR = 0.58, 95% CI:0.45-0.74), and lastly the incomplete vaccination group (OR = 0.64, 95% CI:0.44-0.93). Compared with the complete vaccination-only group, the hybrid immunity (complete vaccination) group mitigated the risk of reinfection by 65% (OR = 0.35, 95% CI:0.27-0.46), and the hybrid immunity (booster vaccination) group mitigated the risk of reinfection by an additional 29% (OR = 0.71, 95% CI:0.61-0.84) compared with the hybrid immunity (complete vaccination) group. The effectiveness of hybrid immunity (incomplete vaccination) in mitigating the risk of reinfection was 37.88% (95% CI, 28.88-46.89%) within 270-364 days, and decreased to 33.23%% (95% CI, 23.80-42.66%) within 365-639 days; whereas, the effectiveness after complete vaccination was 54.36% (95% CI, 50.82-57.90%) within 270-364 days, and the effectiveness of booster vaccination was 73.49% (95% CI, 68.95-78.04%) within 90-119 days. Conclusion Hybrid immunity was significantly more protective than natural or vaccination-induced immunity, and booster doses were associated with enhanced protection against Omicron. Although its protective effects waned over time, vaccination remains a crucial measure for controlling COVID-19. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier, CRD42024539682.
Collapse
Affiliation(s)
- Huiling Zheng
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Shenggen Wu
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Wu Chen
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Shaojian Cai
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Meirong Zhan
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Cailin Chen
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Jiawei Lin
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Zhonghang Xie
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Jianming Ou
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Wenjing Ye
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| |
Collapse
|
350
|
Kamalova D, Akhmetova A, Amirgazin A, Sytnik I, Rudenko V, Yessimkhanova G, Bayesheva D, Yegorov S, Shevtsov A. Genetic Diversity of Human Enterovirus in Kazakhstan, during 2022. Int J Microbiol 2024; 2024:7796913. [PMID: 39224868 PMCID: PMC11368554 DOI: 10.1155/2024/7796913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Enteroviral infection is a common cause of aseptic meningitis, herpangina, and hand, foot, and mouth disease in children. Limited data are available on the enteroviral subtypes associated with hospitalization for these conditions in Kazakhstan. We collected cerebrospinal fluid (CSF) and nasopharyngeal swabs (NSW) from children (N = 152, median age = 8 years) hospitalized with symptoms of aseptic meningitis (AM, N = 139) or herpangina (HA, N = 13) disease. We then genotyped enteroviral subtypes associated with AM (n = 50) and HA (n = 9) using next-generation sequencing (NGS) on the viral protein 1 (VP1), followed up by whole-genome sequencing of the isolated viral species. All identified EVs were species B EV, consisting of five echoviruses (E6, E9, E11, E21, and E25) and three coxsackieviruses (CVA9, CVB3, and CVB5) serotypes within the cohort. The most abundant EVs were CVA9 (38.5%), CVB5 (21.5%), and E6 (13.8%). Most HA samples (6/9) were genotyped with coxsackievirus CVA9, while AM was associated with a variety of both echovirus and coxsackievirus serotypes. The results suggest that coxsackievirus CVA9 may be the dominant serotype circulating in the HA population, while AM is more diverse in terms of circulating echovirus and coxsackievirus serotypes. Further studies are needed to determine the clinical implications of these findings and to investigate potential differences in disease severity or outcomes associated with different EV serotypes.
Collapse
Affiliation(s)
- Dinara Kamalova
- National Center for Biotechnology, Astana, Kazakhstan
- L. N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | | | | | - Igor Sytnik
- National Center for Biotechnology, Astana, Kazakhstan
| | | | | | | | - Sergey Yegorov
- National Center for Biotechnology, Astana, Kazakhstan
- Department of BiologySchool of Sciences and HumanitiesNazarbayev University, Astana, Kazakhstan
| | | |
Collapse
|