51
|
Lassoued N, Yero A, Jenabian MA, Soret R, Pilon N. Efficient enzyme-free method to assess the development and maturation of the innate and adaptive immune systems in the mouse colon. Sci Rep 2024; 14:11063. [PMID: 38744932 PMCID: PMC11094196 DOI: 10.1038/s41598-024-61834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
Researchers who aim to globally analyze the gastrointestinal immune system via flow cytometry have many protocol options to choose from, with specifics generally tied to gut wall layers of interest. To get a clearer idea of the approach we should use on full-thickness colon samples from mice, we first undertook a systematic comparison of three tissue dissociation techniques: two based on enzymatic cocktails and the other one based on manual crushing. Using flow cytometry panels of general markers of lymphoid and myeloid cells, we found that the presence of cell-surface markers and relative cell population frequencies were more stable with the mechanical method. Both enzymatic approaches were associated with a marked decrease of several cell-surface markers. Using mechanical dissociation, we then developed two minimally overlapping panels, consisting of a total of 26 antibodies, for serial profiling of lymphoid and myeloid lineages from the mouse colon in greater detail. Here, we highlight how we accurately delineate these populations by manual gating, as well as the reproducibility of our panels on mouse spleen and whole blood. As a proof-of-principle of the usefulness of our general approach, we also report segment- and life stage-specific patterns of immune cell profiles in the colon. Overall, our data indicate that mechanical dissociation is more suitable and efficient than enzymatic methods for recovering immune cells from all colon layers at once. Additionally, our panels will provide researchers with a relatively simple tool for detailed immune cell profiling in the murine gastrointestinal tract, regardless of life stage or experimental conditions.
Collapse
Affiliation(s)
- Nejia Lassoued
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
| | - Alexis Yero
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Rodolphe Soret
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
52
|
Khales P, Razizadeh MH, Ghorbani S, Moattari A, Sarvari J, Saadati H, Sayyahfar S, Salavatiha Z, Hasanabad MH, Poortahmasebi V, Tavakoli A. Human adenoviruses in children with gastroenteritis: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:478. [PMID: 38724898 PMCID: PMC11084101 DOI: 10.1186/s12879-024-09386-x] [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: 02/06/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024] Open
Abstract
PURPOSE Human adenoviruses (HAdVs) have always been suggested as one of the main causes of gastroenteritis in children. However, no comprehensive report on the global epidemiology of these viruses in pediatric gastroenteritis is available. METHODS A systematic search was conducted to obtain published papers from 2003 to 2023 in three main databases PubMed, Scopus, and Web of Science. RESULTS The estimated global pooled prevalence of HAdV infection in children with gastroenteritis was 10% (95% CI: 9-11%), with a growing trend after 2010. The highest prevalence was observed in Africa (20%, 95% CI: 14-26%). The prevalence was higher in inpatients (11%; 95% CI: 8-13%) and patients aged 5 years old and younger (9%; 95% CI: 7-10%). However, no significant difference was observed between male and female patients (P = 0.63). The most prevalent species was found to be the species F (57%; 95% CI: 41-72%). The most common HAdVs observed in children with gastroenteritis were types 40/41, 38, and 2. Analysis of case-control studies showed an association between HAdV and gastroenteritis in children (OR: 2.28, 95% CI; 1.51-3.44). CONCLUSION This study provided valuable insights into the importance of HAdVs in children with gastroenteritis, especially in hospitalized and younger children. The results can be used in future preventive measurements and the development of effective vaccines.
Collapse
Affiliation(s)
- Pegah Khales
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Razizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Saied Ghorbani
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamal Sarvari
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Saadati
- Department of Epidemiology and Biostatistics, School of Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Shirin Sayyahfar
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Salavatiha
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Haghighi Hasanabad
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
53
|
Bandyopadhyay G, Jehrio MG, Baker C, Bhattacharya S, Misra RS, Huyck HL, Chu C, Myers JR, Ashton J, Polter S, Cochran M, Bushnell T, Dutra J, Katzman PJ, Deutsch GH, Mariani TJ, Pryhuber GS. Bulk RNA sequencing of human pediatric lung cell populations reveals unique transcriptomic signature associated with postnatal pulmonary development. Am J Physiol Lung Cell Mol Physiol 2024; 326:L604-L617. [PMID: 38442187 PMCID: PMC11381037 DOI: 10.1152/ajplung.00385.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
Postnatal lung development results in an increasingly functional organ prepared for gas exchange and pathogenic challenges. It is achieved through cellular differentiation and migration. Changes in the tissue architecture during this development process are well-documented and increasing cellular diversity associated with it are reported in recent years. Despite recent progress, transcriptomic and molecular pathways associated with human postnatal lung development are yet to be fully understood. In this study, we investigated gene expression patterns associated with healthy pediatric lung development in four major enriched cell populations (epithelial, endothelial, and nonendothelial mesenchymal cells, along with lung leukocytes) from 1-day-old to 8-yr-old organ donors with no known lung disease. For analysis, we considered the donors in four age groups [less than 30 days old neonates, 30 days to < 1 yr old infants, toddlers (1 to < 2 yr), and children 2 yr and older] and assessed differentially expressed genes (DEG). We found increasing age-associated transcriptional changes in all four major cell types in pediatric lung. Transition from neonate to infant stage showed highest number of DEG compared with the number of DEG found during infant to toddler- or toddler to older children-transitions. Profiles of differential gene expression and further pathway enrichment analyses indicate functional epithelial cell maturation and increased capability of antigen presentation and chemokine-mediated communication. Our study provides a comprehensive reference of gene expression patterns during healthy pediatric lung development that will be useful in identifying and understanding aberrant gene expression patterns associated with early life respiratory diseases.NEW & NOTEWORTHY This study presents postnatal transcriptomic changes in major cell populations in human lung, namely endothelial, epithelial, mesenchymal cells, and leukocytes. Although human postnatal lung development continues through early adulthood, our results demonstrate that greatest transcriptional changes occur in first few months of life during neonate to infant transition. These early transcriptional changes in lung parenchyma are particularly notable for functional maturation and activation of alveolar type II cell genes.
Collapse
Affiliation(s)
- Gautam Bandyopadhyay
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew G Jehrio
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Cameron Baker
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Ravi S Misra
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Heidie L Huyck
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - ChinYi Chu
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Jason R Myers
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - John Ashton
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - Steven Polter
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew Cochran
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Timothy Bushnell
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Jennifer Dutra
- UR Clinical & Translational Science Institute Informatics, University of Rochester Medical Center, Rochester, New York, United States
| | - Philip J Katzman
- Department of Pathology, University of Rochester Medical Center, Rochester, New York, United States
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital, Seattle, Washington, United States
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
54
|
Fracella M, Mancino E, Nenna R, Virgillito C, Frasca F, D'Auria A, Sorrentino L, Petrarca L, La Regina D, Matera L, Di Mattia G, Caputo B, Antonelli G, Pierangeli A, Viscidi RP, Midulla F, Scagnolari C. Age-related transcript changes in type I interferon signaling in children and adolescents with long COVID. Eur J Immunol 2024; 54:e2350682. [PMID: 38522030 DOI: 10.1002/eji.202350682] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
SARS-CoV-2 typically causes mild symptoms in children, but evidence suggests that persistent immunopathological changes may lead to long COVID (LC). To explore the interplay between LC and innate immunity, we assessed the type I interferon (IFN-I) response in children and adolescents with LC symptoms (LC; n = 28). This was compared with age-matched SARS-CoV-2 recovered participants without LC symptoms (MC; n = 28) and healthy controls (HC; n = 18). We measured the mRNA expression of IFN-I (IFN-α/β/ε/ω), IFN-I receptor (IFNAR1/2), and ISGs (ISG15, ISG56, MxA, IFI27, BST2, LY6E, OAS1, OAS2, OAS3, and MDA5) in PBMCs collected 3-6 months after COVID-19. LC adolescents (12-17 years) had higher transcript levels of IFN-β, IFN-ε, and IFN-ω than HC, whereas LC children (6-11 years) had lower levels than HC. In adolescents, increased levels of IFN-α, IFN-β, and IFN-ω mRNAs were found in the LC group compared with MC, while lower levels were observed in LC children than MC. Adolescents with neurological symptoms had higher IFN-α/β mRNA levels than MC. LC and MC participants showed decreased expression of ISGs and IFNAR1, but increased expression of IFNAR2, than HC. Our results show age-related changes in the expression of transcripts involved in the IFN-I signaling pathway in children and adolescents with LC.
Collapse
Affiliation(s)
- Matteo Fracella
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Enrica Mancino
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Raffaella Nenna
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Chiara Virgillito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Federica Frasca
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandra D'Auria
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Leonardo Sorrentino
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Laura Petrarca
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Domenico La Regina
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Luigi Matera
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Greta Di Mattia
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Beniamino Caputo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Alessandra Pierangeli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| | - Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Fabio Midulla
- Department of Pediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
55
|
Lei H. Quantitative and Longitudinal Assessment of Systemic Innate Immunity in Health and Disease Using a 2D Gene Model. Biomedicines 2024; 12:969. [PMID: 38790931 PMCID: PMC11117654 DOI: 10.3390/biomedicines12050969] [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: 04/01/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Dysregulation of innate immunity is deeply involved in infectious and autoimmune diseases. For a better understanding of pathogenesis and improved management of these diseases, it is of vital importance to implement convenient monitoring of systemic innate immunity. Built upon our previous works on the host transcriptional response to infection in peripheral blood, we proposed a 2D gene model for the simultaneous assessment of two major components of systemic innate immunity, including VirSig as the signature of the host response to viral infection and BacSig as the signature of the host response to bacterial infection. The revelation of dysregulation in innate immunity by this 2D gene model was demonstrated with a wide variety of transcriptome datasets. In acute infection, distinctive patterns of VirSig and BacSig activation were observed in viral and bacterial infection. In comparison, both signatures were restricted to a defined range in the vast majority of healthy adults, regardless of age. In addition, BacSig showed significant elevation during pregnancy and an upward trend during development. In tuberculosis (TB), elevation of BacSig and VirSig was observed in a significant portion of active TB patients, and abnormal BacSig was also associated with a longer treatment course. In cystic fibrosis (CF), abnormal BacSig was observed in a subset of patients, and no overall change in BacSig abnormality was observed after the drug treatment. In systemic sclerosis-associated interstitial lung disease (SSc-ILD), significant elevation of VirSig and BacSig was observed in some patients, and treatment with a drug led to the further deviation of BacSig from the control level. In systemic lupus erythematosus (SLE), positivity for the anti-Ro autoantibody was associated with significant elevation of VirSig in SLE patients, and the additive effect of VirSig/BacSig activation was also observed in SLE patients during pregnancy. Overall, these data demonstrated that the 2D gene model can be used to assess systemic innate immunity in health and disease, with the potential clinical applications including patient stratification, prescription of antibiotics, understanding of pathogenesis, and longitudinal monitoring of treatment response.
Collapse
Affiliation(s)
- Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing 100101, China; ; Tel.: +86-010-8409-7276
- Cunji Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
56
|
Yuan PD, Hu YW, Chen XQ, Chen GY, Pan Y, Lao HY, Liang D. Adalimumab Dose Reduction and Withdrawal in Stable Non-Infectious Pediatric Uveitis: An Open-Label, Prospective, Pilot Study. Ocul Immunol Inflamm 2024:1-8. [PMID: 38652891 DOI: 10.1080/09273948.2024.2343084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
PURPOSES This study investigated the feasibility of adalimumab (ADA) dose reduction and withdrawal strategy in children with stable pediatric non-infectious uveitis (PNIU). METHODS This open-label prospective pilot trial recruited 18 stable PNIU patients (33 eyes) between two and eighteen years old who were treated with standard doses of ADA (20/40 mg every 2 weeks) plus oral methotrexate. The interval of ADA injection was extended to 4 weeks and followed up for 24 weeks. If the uveitis remained stable, ADA was discontinued and followed up for another 24 weeks. ADA was considered successfully stopped if no relapse occurred during this period. The relapse-free survival rate, best corrected visual acuity (BVCA), anterior chamber cell (ACC), vitritis, macular thickness (MT), and serum ADA levels were evaluated. Approval Number: 2021KYPJ201. ClinicalTrials.gov identifier: NCT05155592. RESULTS The relapse-free survival rate was 22.2% (4/18) at 48 weeks. 33.3% (6/18) of patients relapsed when ADA was given every 4 weeks, while 44.5% of patients (8/18) relapsed after ADA was stopped. The four patients successfully withdrawn from ADA were all diagnosed with BD. No statistically significant differences (p > 0.05) were observed in BCVA and MT between baseline and final follow-up. The proportion of ACC and vitritis exhibited an upward trend (p < 0.05) during follow-up. Serum ADA gradually decreased to zero during follow-up in both non-recurrence and recurrence groups. CONCLUSIONS In PNIU children who reached remission for 6 months, ADA dose reduction and withdrawal were associated with a high risk of inflammation recurrence. Timely adjustment of ADA to the last effective dosage frequency can regain control of the inflammation. Detection of ADA serum levels in patients with recurrence may help find the appropriate interval of ADA use.
Collapse
Affiliation(s)
- P D Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
- Department of Ophthalmology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Y W Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
- Department of Ophthalmology, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - X Q Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - G Y Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - Y Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - H Y Lao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| | - D Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
57
|
Beer E, Chowdhury H, Carroll B, Luintel A, van Tulleken C, Longley N. Advising the immunocompromised traveller: a review of immunocompromise at The London Hospital for Tropical Diseases Travel Clinic between 1st April 2019 and 30th April 2020. Trop Dis Travel Med Vaccines 2024; 10:8. [PMID: 38616263 PMCID: PMC11017494 DOI: 10.1186/s40794-024-00217-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/09/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Immunocompromised travellers (ICTs) face greater infectious and non-infectious travel-associated risks than their immunocompetent counterparts. Increasing travel and emergence of novel immunosuppressants poses great challenges for travel medicine practitioners to confidently provide up-to-date evidence-based risk management advice and pre-travel care for ICTs. METHODS We reviewed the records of ICTs attending the London Hospital for Tropical Diseases (HTD) Travel Clinic between 1st April 2019 and 30th April 2020 with the aim to describe demographic and travel characteristics, type, and severity of immunocompromise, the degree of risk associated with intended travel and evaluate travel advice. RESULTS Of the 193 ICTs identified, immunocompromise was due to physiological reasons (42%), chronic infection (17.1%) and immunosuppressive therapy (16.6%). Median age was 38 (range 9 months to 84 years) and male to female ratio 0.75 (83:110). Travel was intended to 80 countries for a median of 16 days (range 2 to 3167), predominantly for leisure (53%), non-medical work (17%) and visiting friends and relatives (12%). Live vaccine safety dominated discussion in the pre-travel consultation. Existing guidelines arguably fell short in dealing with travel risks associated with hyper-specific conditions, targeted immunosuppressants and non-vaccine preventable infections. CONCLUSIONS Our cohort represents a wide spectrum of immunocompromise, for whom we arguably need more measurable ways to approach travel-associated risks. We propose prospective qualitative participatory research to inform our unit of the priorities of ICTs in the pre-travel consultation. We further recommend the formation of a repository of specialists and formulary of complex cases to direct subsequent informative systematic review and prospective risk studies.
Collapse
Affiliation(s)
- Ellen Beer
- University College London Hospital, London, NW1 2BU, UK.
| | | | | | - Akish Luintel
- University College London Hospital, London, NW1 2BU, UK
| | | | - Nicky Longley
- Hospital for Tropical Diseases, University College London Hospital, London, NW1 2BU, UK
- London School of Hygiene & Tropical Medicine, Keppel St, London, WC1E 7HT, UK
| |
Collapse
|
58
|
Charlab R, Leong R, Shord SS, Reaman GH. Pediatric Cancer Drug Development: Leveraging Insights in Cancer Biology and the Evolving Regulatory Landscape to Address Challenges and Guide Further Progress. Cold Spring Harb Perspect Med 2024; 14:a041656. [PMID: 38467448 PMCID: PMC10982696 DOI: 10.1101/cshperspect.a041656] [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: 03/13/2024]
Abstract
The discovery and development of anticancer drugs for pediatric patients have historically languished when compared to both past and recent activity in drug development for adult patients, notably the dramatic spike of targeted and immune-oncology therapies. The reasons for this difference are multifactorial. Recent changes in the regulatory landscape surrounding pediatric cancer drug development and the understanding that some pediatric cancers are driven by genetic perturbations that also drive disparate adult cancers afford new opportunities. The unique cancer-initiating events and dependencies of many pediatric cancers, however, require additional pediatric-specific strategies. Research efforts to unravel the underlying biology of pediatric cancers, innovative clinical trial designs, model-informed drug development, extrapolation from adult data, addressing the unique considerations in pediatric patients, and use of pediatric appropriate formulations, should all be considered for efficient development and dosage optimization of anticancer drugs for pediatric patients.
Collapse
Affiliation(s)
- Rosane Charlab
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Ruby Leong
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Stacy S Shord
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Gregory H Reaman
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
59
|
Nusslock R, Alloy LB, Brody GH, Miller GE. Annual Research Review: Neuroimmune network model of depression: a developmental perspective. J Child Psychol Psychiatry 2024; 65:538-567. [PMID: 38426610 PMCID: PMC11090270 DOI: 10.1111/jcpp.13961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/02/2024]
Abstract
Depression is a serious public health problem, and adolescence is an 'age of risk' for the onset of Major Depressive Disorder. Recently, we and others have proposed neuroimmune network models that highlight bidirectional communication between the brain and the immune system in both mental and physical health, including depression. These models draw on research indicating that the cellular actors (particularly monocytes) and signaling molecules (particularly cytokines) that orchestrate inflammation in the periphery can directly modulate the structure and function of the brain. In the brain, inflammatory activity heightens sensitivity to threats in the cortico-amygdala circuit, lowers sensitivity to rewards in the cortico-striatal circuit, and alters executive control and emotion regulation in the prefrontal cortex. When dysregulated, and particularly under conditions of chronic stress, inflammation can generate feelings of dysphoria, distress, and anhedonia. This is proposed to initiate unhealthy, self-medicating behaviors (e.g. substance use, poor diet) to manage the dysphoria, which further heighten inflammation. Over time, dysregulation in these brain circuits and the inflammatory response may compound each other to form a positive feedback loop, whereby dysregulation in one organ system exacerbates the other. We and others suggest that this neuroimmune dysregulation is a dynamic joint vulnerability for depression, particularly during adolescence. We have three goals for the present paper. First, we extend neuroimmune network models of mental and physical health to generate a developmental framework of risk for the onset of depression during adolescence. Second, we examine how a neuroimmune network perspective can help explain the high rates of comorbidity between depression and other psychiatric disorders across development, and multimorbidity between depression and stress-related medical illnesses. Finally, we consider how identifying neuroimmune pathways to depression can facilitate a 'next generation' of behavioral and biological interventions that target neuroimmune signaling to treat, and ideally prevent, depression in youth and adolescents.
Collapse
Affiliation(s)
- Robin Nusslock
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| | - Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA. USA
| | - Gene H. Brody
- Center for Family Research, University of Georgia, Athens GA, USA
| | - Gregory E. Miller
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| |
Collapse
|
60
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [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: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
61
|
Lee S, Sbihi H, MacIsaac JL, Balshaw R, Ambalavanan A, Subbarao P, Mandhane PJ, Moraes TJ, Turvey SE, Duan Q, Brauer M, Brook JR, Kobor MS, Jones MJ. Persistent DNA Methylation Changes across the First Year of Life and Prenatal NO2 Exposure in a Canadian Prospective Birth Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:47004. [PMID: 38573328 DOI: 10.1289/ehp13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
BACKGROUND Evidence suggests that prenatal air pollution exposure alters DNA methylation (DNAm), which could go on to affect long-term health. It remains unclear whether DNAm alterations present at birth persist through early life. Identifying persistent DNAm changes would provide greater insight into the molecular mechanisms contributing to the association of prenatal air pollution exposure with atopic diseases. OBJECTIVES This study investigated DNAm differences associated with prenatal nitrogen dioxide (NO 2 ) exposure (a surrogate measure of traffic-related air pollution) at birth and 1 y of age and examined their role in atopic disease. We focused on regions showing persistent DNAm differences from birth to 1 y of age and regions uniquely associated with postnatal NO 2 exposure. METHODS Microarrays measured DNAm at birth and at 1 y of age for an atopy-enriched subset of Canadian Health Infant Longitudinal Development (CHILD) study participants. Individual and regional DNAm differences associated with prenatal NO 2 (n = 128 ) were identified, and their persistence at age 1 y were investigated using linear mixed effects models (n = 124 ). Postnatal-specific DNAm differences (n = 125 ) were isolated, and their association with NO 2 in the first year of life was examined. Causal mediation investigated whether DNAm differences mediated associations between NO 2 and age 1 y atopy or wheeze. Analyses were repeated using biological sex-stratified data. RESULTS At birth (n = 128 ), 18 regions of DNAm were associated with NO 2 , with several annotated to HOX genes. Some of these regions were specifically identified in males (n = 73 ), but not females (n = 55 ). The effect of prenatal NO 2 across CpGs within altered regions persisted at 1 y of age. No significant mediation effects were identified. Sex-stratified analyses identified postnatal-specific DNAm alterations. DISCUSSION Regional cord blood DNAm differences associated with prenatal NO 2 persisted through at least the first year of life in CHILD participants. Some differences may represent sex-specific alterations, but replication in larger cohorts is needed. The early postnatal period remained a sensitive window to DNAm perturbations. https://doi.org/10.1289/EHP13034.
Collapse
Affiliation(s)
- Samantha Lee
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Hind Sbihi
- British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia L MacIsaac
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Robert Balshaw
- Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Padmaja Subbarao
- Department of Pediatrics & Translational Medicine, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Piushkumar J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Medicine, USCI University, Kuala Lumpur, Malaysia
| | - Theo J Moraes
- Department of Pediatrics & Translational Medicine, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Stuart E Turvey
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qingling Duan
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- School of Computing, Queen's University, Kingston, Ontario, Canada
| | - Michael Brauer
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Michael S Kobor
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
62
|
Smits HH, Jochems SP. Diverging patterns in innate immunity against respiratory viruses during a lifetime: lessons from the young and the old. Eur Respir Rev 2024; 33:230266. [PMID: 39009407 PMCID: PMC11262623 DOI: 10.1183/16000617.0266-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/16/2024] [Indexed: 07/17/2024] Open
Abstract
Respiratory viral infections frequently lead to severe respiratory disease, particularly in vulnerable populations such as young children, individuals with chronic lung conditions and older adults, resulting in hospitalisation and, in some cases, fatalities. The innate immune system plays a crucial role in monitoring for, and initiating responses to, viruses, maintaining a state of preparedness through the constant expression of antimicrobial defence molecules. Throughout the course of infection, innate immunity remains actively involved, contributing to viral clearance and damage control, with pivotal contributions from airway epithelial cells and resident and newly recruited immune cells. In instances where viral infections persist or are not effectively eliminated, innate immune components prominently contribute to the resulting pathophysiological consequences. Even though both young children and older adults are susceptible to severe respiratory disease caused by various respiratory viruses, the underlying mechanisms may differ significantly. Children face the challenge of developing and maturing their immunity, while older adults contend with issues such as immune senescence and inflammaging. This review aims to compare the innate immune responses in respiratory viral infections across both age groups, identifying common central hubs that could serve as promising targets for innovative therapeutic and preventive strategies, despite the apparent differences in underlying mechanisms.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Jochems
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
63
|
Long J, Zhang J, Chen G, Su X, Qiu B, Dong Q. Pediatric perforated appendicitis diagnosis based on the C-reactive protein/prealbumin ratio. Sci Rep 2024; 14:6729. [PMID: 38509094 PMCID: PMC10954718 DOI: 10.1038/s41598-024-55108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Pediatric perforated appendicitis, prone to multiple complications, necessitates identifying potential serum biomarkers for early diagnosis and intervention. A cross-sectional study was conducted on patients under 16 with acute appendicitis, admitted to Hainan Women and Children's Medical Center from January 2019 to July 2023. The patients were categorized into perforated and non-perforated groups. Among the 313 included patients, 106 (33.87%, 95% CI 28.59-39.14%) developed perforation. The C-reactive protein to prealbumin ratio (CPA) showed a significant difference between the perforated and non-perforated groups [6.63 (2.9-13.02) vs. 0.7 (0.11-2.18), p < 0.001]. The AUC of CPA on the ROC curve was 0.691 (95% CI 0.513-0.869, p = 0.084) in patients under 4. In patients aged 4-9, the sensitivity of CPA > 3 predicting perforation was 76.2%, with a specificity of 81.6%, and an AUC of 0.816 (95% CI 0.747-0.886, p < 0.001). For patients aged 9-16, the sensitivity of CPA > 2.2 predicting perforation was 85%, with a specificity of 85.7%, and an AUC of 0.919 (95% CI 0.859-0.979, p < 0.001). CPA > 3 and CPA > 2.2 can predict perforated appendicitis in patients aged 4-9 and 9-16, respectively.
Collapse
Affiliation(s)
- Junshan Long
- Department of General Surgery, Hainan Women and Children's Medical Center, Changbin Road, Haikou, Hainan, China
| | - Jing Zhang
- Department of General Surgery, Hainan Women and Children's Medical Center, Changbin Road, Haikou, Hainan, China
| | - Gong Chen
- Department of General Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoxia Su
- Department of General Surgery, Hainan Women and Children's Medical Center, Changbin Road, Haikou, Hainan, China
| | - Baowei Qiu
- Department of General Surgery, Hainan Women and Children's Medical Center, Changbin Road, Haikou, Hainan, China
| | - Qi Dong
- Department of General Surgery, Hainan Women and Children's Medical Center, Changbin Road, Haikou, Hainan, China.
| |
Collapse
|
64
|
Allotey-Babington GL, Akwo Kretchy I, Atiapa Asiedu E, Kelly Amuakwa M, Akwele Seaneke O, Ankrah D, Kwadwo Somuah A, Dei Owusu-Nyamekye A, Owusu-Ansah S, Kwame Effah P, Debrah J, Acheampomaa Nai E, Owusu E, Lamptey W, Gyekye IJA, Nettey H. Prevalence, Scope and Quality of Extemporaneous Medications in Selected Healthcare Facilities and Implications for Pharmacy Practice. Innov Pharm 2024; 15:10.24926/iip.v15i1.5971. [PMID: 38779112 PMCID: PMC11107970 DOI: 10.24926/iip.v15i1.5971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Background: Extemporaneous compounding is the preparation of medicines for individual patients when no commercially available authorized form exists. Unlike registered medications, these products are not subjected to various tests for quality by Regulatory Authorities. Data on compounded medications in Ghana is currently inadequate or unavailable. There is the need to collate data that can be used to influence policy and to regulate preparation of extemporaneous products. Aim: To establish the prevalence, scope and quality of extemporaneously compounded medicines at selected hospitals in Accra, Ghana. Methodology: Prescriptions presented at the pharmacies in selected hospitals were reviewed to determine the requests that needed to be extemporaneously prepared as well as the prevalence and the scope of formulations. Three of the most frequently compounded medications were procured and subjected to microbial contamination tests using the pour plate method followed by differential tests if microbes were present. Content analysis of the active ingredients was determined using High Performance Liquid Chromatography (HPLC). Results: 641 requests comprising 49 different extemporaneous products were collated from the hospitals studied. Hydroxyurea, furosemide and spironolactone suspensions were the three most frequently prescribed. Patients aged from 0-2 years had majority of the prescriptions. Conclusion: A population of patients still exist who depend on compounding for their drug needs. 49 different formulations were prepared at one of the hospitals visited. Samples of products analyzed were of good quality.
Collapse
Affiliation(s)
| | - Irene Akwo Kretchy
- Department of Pharmacy Practice and clinical Pharmacy, School of Pharmacy University of Ghana, P.O. Box LG 43 Legon, Ghana
| | - Esther Atiapa Asiedu
- Department of Pharmaceutics and Microbiology, School of Pharmacy University of Ghana, P.O. Box LG 43 Legon, Ghana
| | - Maxine Kelly Amuakwa
- Department of Pharmaceutics and Microbiology, School of Pharmacy University of Ghana, P.O. Box LG 43 Legon, Ghana
| | | | - Daniel Ankrah
- Korle Bu Teaching Hospital P.O. Box 77, Korle Bu Ghana
| | | | | | | | | | | | | | - Emmanuel Owusu
- Greater Accra Regional Hospital P.O. Box GP 473 Accra, Ghana
| | - William Lamptey
- Princess Marie Children’s Hospital P.O. Box GP 122 Accra, Ghana
| | - Isaac Julius Asiedu Gyekye
- Department of Pharmacology and Toxicology, School of Pharmacy University of Ghana, P.O. Box LG 43 Legon, Ghana
| | - Henry Nettey
- Department of Pharmaceutics and Microbiology, School of Pharmacy University of Ghana, P.O. Box LG 43 Legon, Ghana
| |
Collapse
|
65
|
Jamali Z, Mohammadpour N, Sinaei R, Jafari M, Sabzevari F, Hasannejad M. The footprint of SARS-COV-2 infection in neonatal late sepsis. BMC Pediatr 2024; 24:184. [PMID: 38491449 PMCID: PMC10943770 DOI: 10.1186/s12887-024-04665-7] [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: 06/15/2023] [Accepted: 02/22/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Predicting and finding the viral agents responsible for neonatal late-sepsis has always been challenging. METHOD In this cross-sectional study, which has been done from September 2020 to December 2022, 145 hospitalized neonates suspected to late-onset sepsis alongside routine sepsis workup, were also evaluated for severe acute respiratory syndrome-coronavirus-2 (SARS-COV-2) infection, by nasopharyngeal real-time polymerase chain reaction (RT-PCR) or serological tests. RESULT 145 neonates including 81 girls and 64 boys with a mean age of 12.3 ± 5.9 days and an average hospitalization stay of 23.1 ± 15.4 days were enrolled in the study. While 76.6% of them had negative bacterial culture, 63 patients (43.4%) showed evidence of SARS-COV-2 infection in RT-PCR or serology tests. None of the underlying factors including gender, age, and laboratory investigation had a significant relationship with SARS-COV-2 infection. Similarly, the outcomes of death and length of hospitalization were not different between the two groups with positive and negative SARS-COV-2 RT-PCR (P < 0.05). There was only a significant relationship between radiological changes including reticulonodular pattern, consolidation, pleural effusion, and different types of infiltrations and SARS-COV2 infection. CONCLUSION Considering the widespread of coronavirus disease 2019 (COVID-19) in newborns, it seems logical to investigate the SARS-COV-2 infection in late-sepsis.
Collapse
Affiliation(s)
- Zahra Jamali
- Department of Pediatrics, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Najmeh Mohammadpour
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Sinaei
- Department of Pediatrics, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Maedeh Jafari
- Department of Pediatrics, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sabzevari
- Department of Pediatrics, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hasannejad
- Department of Laboratory Sciences, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| |
Collapse
|
66
|
Merz MP, Seal SV, Grova N, Mériaux S, Guebels P, Kanli G, Mommaerts E, Nicot N, Kaoma T, Keunen O, Nazarov PV, Turner JD. Early-life influenza A (H1N1) infection independently programs brain connectivity, HPA AXIS and tissue-specific gene expression profiles. Sci Rep 2024; 14:5898. [PMID: 38467724 PMCID: PMC10928197 DOI: 10.1038/s41598-024-56601-5] [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: 09/22/2023] [Accepted: 03/08/2024] [Indexed: 03/13/2024] Open
Abstract
Early-life adversity covers a range of physical, social and environmental stressors. Acute viral infections in early life are a major source of such adversity and have been associated with a broad spectrum of later-life effects outside the immune system or "off-target". These include an altered hypothalamus-pituitary-adrenal (HPA) axis and metabolic reactions. Here, we used a murine post-natal day 14 (PND 14) Influenza A (H1N1) infection model and applied a semi-holistic approach including phenotypic measurements, gene expression arrays and diffusion neuroimaging techniques to investigate HPA axis dysregulation, energy metabolism and brain connectivity. By PND 56 the H1N1 infection had been resolved, and there was no residual gene expression signature of immune cell infiltration into the liver, adrenal gland or brain tissues examined nor of immune-related signalling. A resolved early-life H1N1 infection had sex-specific effects. We observed retarded growth of males and altered pre-stress (baseline) blood glucose and corticosterone levels at PND42 after the infection was resolved. Cerebral MRI scans identified reduced connectivity in the cortex, midbrain and cerebellum that were accompanied by tissue-specific gene expression signatures. Gene set enrichment analysis confirmed that these were tissue-specific changes with few common pathways. Early-life infection independently affected each of the systems and this was independent of HPA axis or immune perturbations.
Collapse
Affiliation(s)
- Myriam P Merz
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de Université, L-4365, Esch-Sur-Alzette, Luxembourg
- Central Biobank Charité, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Snehaa V Seal
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 2 Avenue de Université, L-4365, Esch-Sur-Alzette, Luxembourg
| | - Nathalie Grova
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg
- Inserm U1256, NGERE, Nutrition-Génétique Et Exposition Aux Risques Environnementaux, Université de Lorraine, 54000, Nancy, France
| | - Sophie Mériaux
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg
| | - Pauline Guebels
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg
| | - Georgia Kanli
- In Vivo Imaging Platform, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
- Translational Radiomics, Department of Cancer Research, Luxembourg Institute of Health, 1526, Luxembourg, Luxembourg
| | - Elise Mommaerts
- LuxGen Genome Center, Laboratoire National de Santé, Luxembourg Institute of Health, 3555, Dudelange, Luxembourg
| | - Nathalie Nicot
- LuxGen Genome Center, Laboratoire National de Santé, Luxembourg Institute of Health, 3555, Dudelange, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Data Integration and Analysis Unit, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Olivier Keunen
- In Vivo Imaging Platform, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
- Translational Radiomics, Department of Cancer Research, Luxembourg Institute of Health, 1526, Luxembourg, Luxembourg
| | - Petr V Nazarov
- Bioinformatics Platform, Data Integration and Analysis Unit, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
- Multiomics Data Science Research Group, Department of Cancer Research, Luxembourg Institute of Health, 1445, Strassen, Luxembourg
| | - Jonathan D Turner
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 Rue Henri Koch, 4354, Esch-Sur-Alzette, Luxembourg.
| |
Collapse
|
67
|
Wang Y, Gao Y, Shi C, Shen Y, Lu M, Sha D, Chen Y, Zhu D, Shi P. Prevalence, Clinical Features, and Genotypes of Norovirus-Associated Diarrhea in Wuxi, China, 2013-2020. Am J Trop Med Hyg 2024; 110:569-575. [PMID: 38266292 PMCID: PMC10919189 DOI: 10.4269/ajtmh.23-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/30/2023] [Indexed: 01/26/2024] Open
Abstract
Norovirus (NoV) is a common pathogen that can cause infectious diarrhea. This study aimed to determine the prevalence, clinical features, and genotypes of NoV-associated diarrhea in Wuxi, China. A total of 4,416 stool samples were collected from patients with diarrhea at enteric disease clinics of sentinel hospitals in Wuxi from February 1, 2013 to December 31, 2020. Univariate and Akaike information criterion stepwise logistic regression were used to identify differences as integrated within a clinical setting (NoV positive [+] versus NoV negative [-], NoV+ versus rotavirus [RV]+, NoV+ versus bacteria+, genogroup [G] I and GII genotypes). Norovirus was detected in 9.85% of stool samples, which was greater than other tested pathogens. Excluding coinfection of NoV and other viruses or bacteria, patients infected with NoV had a lower chance of acquiring the virus in summer (P < 0.001; odds ratio [OR], 0.257; 95% CI, 0.189-0.36) when compared with patients without NoV. Patients with diarrhea infected with NoV featured nausea and vomiting (P < 0.001; OR, 2.297, 95% CI, 1.85-2.86) and loose stools (P = 0.006; OR, 2.247; 95% CI, 1.30-4.10), but less abdominal cramping (P = 0.001; OR, 0.676; 95% CI, 0.54-0.84). Patients infected with RV (P < 0.001; OR, 0.413; 95% CI, 0.25-0.68) or bacteria (P < 0.001; OR, 0.422; 95% CI, 0.26-0.67) were more vulnerable to fever than those infected with NoV. A total of 379 GII strains were detected concomitant with 48 GI strains, and there was a seasonal difference between the GI and GII genotypes. Strengthening pathogen detection for infectious diarrhea was helpful for understanding the epidemiological characteristics of infections with NoV and, potentially, for preventing disease outbreaks.
Collapse
Affiliation(s)
- Yan Wang
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Yumeng Gao
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Chao Shi
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Yuan Shen
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Mingyan Lu
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Dan Sha
- Microbiological Laboratory, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Yujun Chen
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Ding Zhu
- Department of Disinfection and Vector Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Ping Shi
- Department of Acute Infectious Disease Prevention and Control, The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| |
Collapse
|
68
|
Cai W, Köndgen S, Tolksdorf K, Dürrwald R, Schuler E, Biere B, Schweiger B, Goerlitz L, Haas W, Wolff T, Buda S, Reiche J. Atypical age distribution and high disease severity in children with RSV infections during two irregular epidemic seasons throughout the COVID-19 pandemic, Germany, 2021 to 2023. Euro Surveill 2024; 29:2300465. [PMID: 38551098 PMCID: PMC10979527 DOI: 10.2807/1560-7917.es.2024.29.13.2300465] [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: 08/28/2023] [Accepted: 02/01/2024] [Indexed: 04/01/2024] Open
Abstract
BackgroundNon-pharmaceutical interventions (NPIs) during the COVID-19 pandemic affected respiratory syncytial virus (RSV) circulation worldwide.AimTo describe, for children aged < 5 years, the 2021 and 2022/23 RSV seasons in Germany.MethodsThrough data and 16,754 specimens from outpatient sentinel surveillance, we investigated RSV seasonality, circulating lineages, and affected children's age distributions in 2021 and 2022/23. Available information about disease severity from hospital surveillance was analysed for patients with RSV-specific diagnosis codes (n = 13,104). Differences between RSV seasons were assessed by chi-squared test and age distributions trends by Mann-Kendall test.ResultsRSV seasonality was irregular in 2021 (weeks 35-50) and 2022/23 (weeks 41-3) compared to pre-COVID-19 2011/12-2019/20 seasons (median weeks 51-12). RSV positivity rates (RSV-PR) were higher in 2021 (40% (522/1,291); p < 0.001) and 2022/23 (30% (299/990); p = 0.005) than in prior seasons (26% (1,430/5,511)). Known globally circulating RSV-A (lineages GA2.3.5 and GA2.3.6b) and RSV-B (lineage GB5.0.5a) strains, respectively, dominated in 2021 and 2022/23. In 2021, RSV-PRs were similar in 1 - < 2, 2 - < 3, 3 - < 4, and 4 - < 5-year-olds. RSV hospitalisation incidence in 2021 (1,114/100,000, p < 0.001) and in 2022/23 (1,034/100,000, p < 0.001) was approximately double that of previous seasons' average (2014/15-2019/20: 584/100,000). In 2022/23, proportions of RSV patients admitted to intensive care units rose (8.5% (206/2,413)) relative to pre-COVID-19 seasons (6.8% (551/8,114); p = 0.004), as did those needing ventilator support (6.1% (146/2,413) vs 3.8% (310/8,114); p < 0.001).ConclusionsHigh RSV-infection risk in 2-4-year-olds in 2021 and increased disease severity in 2022/23 possibly result from lower baseline population immunity, after NPIs diminished exposure to RSV.
Collapse
Affiliation(s)
- Wei Cai
- Unit 36, Respiratory Infections, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Sophie Köndgen
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, Consultant Laboratory for RSV, PIV and HMPV, Robert Koch Institute, Berlin, Germany
| | - Kristin Tolksdorf
- Unit 36, Respiratory Infections, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Ralf Dürrwald
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, Consultant Laboratory for RSV, PIV and HMPV, Robert Koch Institute, Berlin, Germany
| | | | - Barbara Biere
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
| | - Brunhilde Schweiger
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
| | - Luise Goerlitz
- Unit 36, Respiratory Infections, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Walter Haas
- Unit 36, Respiratory Infections, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, Consultant Laboratory for RSV, PIV and HMPV, Robert Koch Institute, Berlin, Germany
| | - Silke Buda
- Unit 36, Respiratory Infections, Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Janine Reiche
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, National Influenza Centre, Robert Koch Institute, Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Department of Infectious Diseases, Consultant Laboratory for RSV, PIV and HMPV, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
69
|
Waterlow NR, Cooper BS, Robotham JV, Knight GM. Antimicrobial resistance prevalence in bloodstream infection in 29 European countries by age and sex: An observational study. PLoS Med 2024; 21:e1004301. [PMID: 38484006 PMCID: PMC10939247 DOI: 10.1371/journal.pmed.1004301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/22/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Antibiotic usage, contact with high transmission healthcare settings as well as changes in immune system function all vary by a patient's age and sex. Yet, most analyses of antimicrobial resistance (AMR) ignore demographic indicators and provide only country-level resistance prevalence values. This study aimed to address this knowledge gap by quantifying how resistance prevalence and incidence of bloodstream infection (BSI) varied by age and sex across bacteria and antibiotics in Europe. METHODS AND FINDINGS We used patient-level data collected as part of routine surveillance between 2015 and 2019 on BSIs in 29 European countries from the European Antimicrobial Resistance Surveillance Network (EARS-Net). A total of 6,862,577 susceptibility results from isolates with age, sex, and spatial information from 944,520 individuals were used to characterise resistance prevalence patterns for 38 different bacterial species and antibiotic combinations, and 47% of these susceptibility results were from females, with a similar age distribution in both sexes (mean of 66 years old). A total of 349,448 isolates from 2019 with age and sex metadata were used to calculate incidence. We fit Bayesian multilevel regression models by country, laboratory code, sex, age, and year of sample to quantify resistant prevalence and provide estimates of country-, bacteria-, and drug-family effect variation. We explore our results in greater depths for 2 of the most clinically important bacteria-antibiotic combinations (aminopenicillin resistance in Escherichia coli and methicillin resistance in Staphylococcus aureus) and present a simplifying indicative index of the difference in predicted resistance between old (aged 100) and young (aged 1). At the European level, we find distinct patterns in resistance prevalence by age. Trends often vary more within an antibiotic family, such as fluroquinolones, than within a bacterial species, such as Pseudomonas aeruginosa. Clear resistance increases by age for methicillin-resistant Staphylococcus aureus (MRSA) contrast with a peak in resistance to several antibiotics at approximately 30 years of age for P. aeruginosa. For most bacterial species, there was a u-shaped pattern of infection incidence with age, which was higher in males. An important exception was E. coli, for which there was an elevated incidence in females between the ages of 15 and 40. At the country-level, subnational differences account for a large amount of resistance variation (approximately 38%), and there are a range of functional forms for the associations between age and resistance prevalence. For MRSA, age trends were mostly positive, with 72% (n = 21) of countries seeing an increased resistance between males aged 1 and 100 years and a greater change in resistance in males. This compares to age trends for aminopenicillin resistance in E. coli which were mostly negative (males: 93% (n = 27) of countries see decreased resistance between those aged 1 and 100 years) with a smaller change in resistance in females. A change in resistance prevalence between those aged 1 and 100 years ranged up to 0.51 (median, 95% quantile of model simulated prevalence using posterior parameter ranges 0.48, 0.55 in males) for MRSA in one country but varied between 0.16 (95% quantile 0.12, 0.21 in females) to -0.27 (95% quantile -0.4, -0.15 in males) across individual countries for aminopenicillin resistance in E. coli. Limitations include potential bias due to the nature of routine surveillance and dependency of results on model structure. CONCLUSIONS In this study, we found that the prevalence of resistance in BSIs in Europe varies substantially by bacteria and antibiotic over the age and sex of the patient shedding new light on gaps in our understanding of AMR epidemiology. Future work is needed to determine the drivers of these associations in order to more effectively target transmission and antibiotic stewardship interventions.
Collapse
Affiliation(s)
- Naomi R. Waterlow
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, EPH, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ben S. Cooper
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie V. Robotham
- NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in Partnership with the UK Health Security Agency, Oxford, United Kingdom
| | - Gwenan Mary Knight
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, EPH, London School of Hygiene and Tropical Medicine, London, United Kingdom
- AMR Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
70
|
Kim MS, Kim JH, Ryu S, Lee SW, Yon DK, Kim E, Koyanagi A, Dragioti E, Shin JI, Smith L. Comparative efficacy and optimal duration of first-line antibiotic regimens for acute otitis media in children and adolescents: a systematic review and network meta-analysis of 89 randomized clinical trials. World J Pediatr 2024; 20:219-229. [PMID: 37016201 DOI: 10.1007/s12519-023-00716-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/05/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Antibiotic use for acute otitis media (AOM) is one of the major sources of antimicrobial resistance. However, the effective minimal antibiotic duration for AOM remains unclear. Moreover, guidelines often recommend broad ranges (5-10 days) of antibiotic use, yet the clinical impact of such a wide window has not been assessed. METHODS We systematically searched PubMed/MEDLINE, Embase, Scopus, Web of Science, and Cochrane Library from database inception to 6 October 2021. Network meta-analysis was conducted on randomized controlled trials that assessed antibiotic treatment for AOM in children (PROSPERO CRD42020196107). RESULTS For amoxicillin and amoxicillin-clavulanate, 7-day regimens were noninferior to 10-day regimens in clinical responses [amoxicillin: risk ratio (RR) 0.919 (95% CI 0.820-1.031), amoxicillin-clavulanate: RR 1.108 (0.957-1.282)], except for ≤ 2 years. For the third-generation cephalosporins, 7-day and 10-day regimens had similar clinical responses compared to placebo [7-day: RR 1.420 (1.190-1.694), 10-day: RR 1.238 (1.125-1.362) compared to placebo]. However, 5-day regimens of amoxicillin-clavulanate and third-generation cephalosporins were inferior to 10-day regimens. Compared to amoxicillin, a shorter treatment duration was tolerable with amoxicillin-clavulanate. CONCLUSIONS Our findings indicated that 10 days of antibiotic use may be unnecessarily long, while the treatment duration should be longer than 5 days. Otherwise, 5-day regimens would be sufficient for a modest treatment goal. Our findings revealed that the current wide range of recommended antibiotic durations may have influenced the clinical outcome of AOM, and a narrower antibiotic duration window should be re-established.
Collapse
Affiliation(s)
- Min Seo Kim
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jae Han Kim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seohyun Ryu
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Won Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Eunyoung Kim
- Evidence-Based Research Laboratory, Department of Clinical Pharmacy and Pharmaceutical Care, College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ai Koyanagi
- Parc Sanitari Sant Joan de Déu/CIBERSAM/ISCIII, Universitat de Barcelona, Fundació Sant Joan de Déu, Sant Boi de Llobregat, Barcelona, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain
| | - Elena Dragioti
- Pain and Rehabilitation Centre, and Department of Medical and Health Sciences, Linköping University, 581 85, Linköping, Sweden
- Research Laboratory Psychology of Patients, Families & Health Professionals, Department of Nursing, School of Health Sciences, University of Ioannina, Ioannina, 45500, Greece
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Yonsei-ro 50, Seodaemun-gu, 8044, Seoul, 03722, Republic of Korea.
| | - Lee Smith
- Centre for Health, Performance, and Wellbeing, Anglia Ruskin University, Cambridge, CB1 1PT, UK
| |
Collapse
|
71
|
Chen D, Havelaar AH, Platts-Mills JA, Yang Y. Acquisition and clearance dynamics of Campylobacter spp. in children in low- and middle-income countries. Epidemics 2024; 46:100749. [PMID: 38367286 PMCID: PMC10944168 DOI: 10.1016/j.epidem.2024.100749] [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: 05/28/2023] [Revised: 11/08/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
The prevalence of Campylobacter infection is generally high among children in low- and middle-income countries (LMIC), but the dynamics of its acquisition and clearance are understudied. We aim to quantify this process among children under two years old in eight LMIC using a statistical modeling approach, leveraging enzyme-immunoassay-based Campylobacter genus data and quantitative-PCR-based Campylobacter jejuni/coli data from the MAL-ED study. We developed a Markov model to compare the dynamics of acquisition and clearance of Campylobacter across countries and to explore the effect of antibiotic usage on Campylobacter clearance. Clearance rates were generally higher than acquisition rates, but their magnitude and temporal pattern varied across countries. For C. jejuni/coli, clearance was faster than acquisition throughout the two years at all sites. For Campylobacter spp., the acquisition rate either exceeded or stayed very close to the clearance rate after the first half year in Bangladesh, Pakistan and Tanzania, leading to high prevalence. Bangladesh had the shortest (28 and 57 days) while Brazil had the longest (328 and 306 days) mean times from last clearance to acquisition for Campylobacter spp. and C. jejuni/coli, respectively. South Africa had the shortest (10 and 8 days) while Tanzania had the longest (53 and 41 days) mean times to clearance for Campylobacter spp. and C. jejuni/col, respectively. The use of Macrolide accelerated clearance of C. jejuni/coli in Bangladesh and Peru and of Campylobacter spp. in Bangladesh and Pakistan. Fluoroquinolone showed statistically meaningful effects only in Bangladesh but for both Campylobacter groups. Higher prevalence of Campylobacter infection was mainly driven by a high acquisition rate that was close to or surpassing the clearance rate. Acquisition rate usually peaked in 11-17 months of age, indicating the importance of targeting the first year of life for effective interventions to reduce exposures.
Collapse
Affiliation(s)
- Dehao Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA; Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA; Department of Epidemiology, Emory University, Atlanta, GA, USA
| | - Arie H Havelaar
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA; Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA; Global Food Systems Institute, University of Florida, Gainesville, FL, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia Health System, Charlottesville, VA, USA
| | - Yang Yang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
72
|
Edstorp J, Rossides M, Ahlqvist E, Rasouli B, Tuomi T, Carlsson S. Does a prior diagnosis of infectious disease confer an increased risk of latent autoimmune diabetes in adults? Diabetes Metab Res Rev 2024; 40:e3758. [PMID: 38103209 DOI: 10.1002/dmrr.3758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 12/18/2023]
Abstract
AIMS Infections are proposed risk factors for type 1 diabetes in children. We examined whether a diagnosis of infectious disease also confers an increased risk of latent autoimmune diabetes in adults (LADA). MATERIALS AND METHODS We used data from a population-based Swedish case-control study with incident cases of LADA (n = 597) and matched controls (n = 2386). The history of infectious disease was ascertained through national and regional patient registers. We estimated adjusted odds ratios (OR) with 95% confidence intervals for ≥1 respiratory (any/upper/lower), gastrointestinal, herpetic, other or any infectious disease episode, or separately, for 1 and ≥2 infectious disease episodes, within 0-1, 1-3, 3-5 and 5-10 years before LADA diagnosis/matching. Stratified analyses were performed on the basis of HLA risk genotypes and Glutamic acid decarboxylase antibodies (GADA) levels. RESULTS Individuals who developed LADA did not have a higher prevalence of infectious disease 1-10 years before diabetes diagnosis. For example, OR was estimated at 0.87 (0.66, 1.14) for any versus no respiratory infectious disease within 1-3 years. Similar results were seen for LADA with high-risk HLA genotypes (OR 0.95 [0.64, 1.42]) or high GADA levels (OR 1.10 [0.79, 1.55]), ≥2 episodes (OR 0.89 [0.56, 1.40]), and in infections treated using antibiotics (OR 1.03 [0.73, 1.45]). The only significant association was observed with lower respiratory disease the year preceding LADA diagnosis (OR 1.67 [1.06, 2.64]). CONCLUSIONS Our findings do not support the idea that exposure to infections increases the risk of LADA. A higher prevalence of respiratory infection in the year before LADA diagnosis could reflect increased susceptibility to infections due to hyperglycemia.
Collapse
Affiliation(s)
- Jessica Edstorp
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marios Rossides
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Ahlqvist
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, Malmö, Sweden
| | - Bahareh Rasouli
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tiinamaija Tuomi
- Department of Clinical Sciences in Malmö, Clinical Research Centre, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland, Helsinki University, Helsinki, Finland
- Division of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
- Research Program for Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
73
|
Adeniyi-Ipadeola GO, Hankins JD, Kambal A, Zeng XL, Patil K, Poplaski V, Bomidi C, Nguyen-Phuc H, Grimm SL, Coarfa C, Stossi F, Crawford SE, Blutt SE, Speer AL, Estes MK, Ramani S. Infant and Adult Human Intestinal Enteroids are Morphologically and Functionally Distinct. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.19.541350. [PMID: 37292968 PMCID: PMC10245709 DOI: 10.1101/2023.05.19.541350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background & Aims Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. Methods We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We validated differences in key pathways through functional studies and determined if these cultures recapitulate known features of the infant intestinal epithelium. Results RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. Conclusions HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex-vivo model to advance studies of infant-specific diseases and drug discovery for this population.
Collapse
Affiliation(s)
| | - Julia D. Hankins
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Amal Kambal
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core
| | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core
| | - Ketki Patil
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Victoria Poplaski
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Hoa Nguyen-Phuc
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Sandra L. Grimm
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
- Golf Coast Consortium Center for Advanced Microscopy and Image Informatics, Houston, TX
| | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core
| | - Allison L. Speer
- Department of Pediatric Surgery, The University of Texas Health Science Center, Houston, TX
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
- Texas Medical Center Digestive Diseases Center Gastrointestinal Experimental Model Systems (GEMS) Core
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
74
|
Nour-Eldine W, Manaph NPA, Ltaief SM, Abdel Aati N, Mansoori MH, Al Abdulla S, Al-Shammari AR. Discovery of a novel cytokine signature for the diagnosis of autism spectrum disorder in young Arab children in Qatar. Front Psychiatry 2024; 15:1333534. [PMID: 38414501 PMCID: PMC10896998 DOI: 10.3389/fpsyt.2024.1333534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/22/2024] [Indexed: 02/29/2024] Open
Abstract
Background Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder characterized by impaired social interaction and communication and the occurrence of stereotyped and repetitive behaviors. Several studies have reported altered cytokine profiles in ASD and hence may serve as potential diagnostic biomarkers of the disorder. This study aims to identify diagnostic biomarkers for ASD in a well-defined study cohort in Qatar. Methods We measured the protein levels of 45 cytokines in the plasma samples of age- and gender-matched children (2-4 years) with ASD (n = 100) and controls (n = 60) using a Luminex multiplex assay. We compared the differences in the levels of these cytokines between the two study groups and then fitted the significantly altered cytokines into a logistic regression model to examine their diagnostic potential for ASD. Results We found elevated levels of IFN-γ, FGF-2, IL-1RA, and IL-13 and reduced levels of eotaxin, HGF, IL-1 alpha, IL-22, IL-9, MCP-1, SCF, SDF-1 alpha, VEGFA, and IP-10 in the plasma of children with ASD compared to controls. Furthermore, we observed that elevated levels of IFN-γ (odds ratio (OR) = 1.823; 95% (confidence interval) CI = 1.206, 2.755; p = 0.004) and FGF-2 (OR = 2.528; 95% CI = 1.457, 4.385; p < 0.001) were significantly associated with increased odds of ASD, whereas reduced levels of eotaxin (OR = 0.350; 95% CI = 0.160, 0.765; p = 0.008) and HGF (OR = 0.220; 95% CI = 0.070, 0.696; p = 0.010) were significantly associated with lower odds of ASD relative to controls. The combination of these four cytokines revealed an area under the curve (ROC-AUC) of 0.829 (95% CI = 0.767, 0.891; p < 0.001), which demonstrates the diagnostic accuracy of the four-cytokine signature. Conclusions Our results identified a panel of cytokines that could discriminate between children with ASD and controls in Qatar. In addition, our findings support the predominance of a Th1 immune phenotype in ASD children and emphasize the need to validate these results in larger populations.
Collapse
Affiliation(s)
- Wared Nour-Eldine
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | | | - Samia M Ltaief
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Nazim Abdel Aati
- Child Development Center, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Samya Al Abdulla
- Department of Operations, Primary Health Care Corporation, Doha, Qatar
| | - Abeer R Al-Shammari
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
75
|
Carlberg C, Velleuer E. Vitamin D and Aging: Central Role of Immunocompetence. Nutrients 2024; 16:398. [PMID: 38337682 PMCID: PMC10857325 DOI: 10.3390/nu16030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
The pro-hormone vitamin D3 is an important modulator of both innate and adaptive immunity since its biologically active metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) regulates via the transcription factor VDR (vitamin D receptor) the epigenome and transcriptome of human immune cells and controls in this way the expression of hundreds of vitamin D target genes. Since the myeloid linage of hematopoiesis is epigenetically programmed by VDR in concert with the pioneer factors PU.1 (purine-rich box 1) and CEBPα (CCAAT/enhancer binding protein α), monocytes, macrophages, and dendritic cells are the most vitamin D-sensitive immune cell types. The central role of the immune system in various aging-related diseases suggests that immunocompetence describes not only the ability of an individual to resist pathogens and parasites but also to contest non-communicative diseases and the process of aging itself. In this review, we argue that the individual-specific responsiveness to vitamin D relates to a person's immunocompetence via the epigenetic programming function of VDR and its ligand 1,25(OH)2D3 during hematopoiesis as well as in the periphery. This may provide a mechanism explaining how vitamin D protects against major common diseases and, in parallel, promotes healthy aging.
Collapse
Affiliation(s)
- Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, PL-10-748 Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Eunike Velleuer
- Department for Cytopathology, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany;
- Department for Pediatric Hemato-Oncology, Helios Children’s Hospital, D-47805 Krefeld, Germany
| |
Collapse
|
76
|
Nziza N, Deng Y, Wood L, Dhanoa N, Dulit-Greenberg N, Chen T, Kane AS, Swank Z, Davis JP, Demokritou M, Chitnis AP, Fasano A, Edlow AG, Jain N, Horwitz BH, McNamara RP, Walt DR, Lauffenburger DA, Julg B, Shreffler WG, Alter G, Yonker LM. Humoral profiles of toddlers and young children following SARS-CoV-2 mRNA vaccination. Nat Commun 2024; 15:905. [PMID: 38291080 PMCID: PMC10827750 DOI: 10.1038/s41467-024-45181-7] [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: 03/28/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Although young children generally experience mild symptoms following infection with SARS-CoV-2, severe acute and long-term complications can occur. SARS-CoV-2 mRNA vaccines elicit robust immunoglobulin profiles in children ages 5 years and older, and in adults, corresponding with substantial protection against hospitalizations and severe disease. Whether similar immune responses and humoral protection can be observed in vaccinated infants and young children, who have a developing and vulnerable immune system, remains poorly understood. To study the impact of mRNA vaccination on the humoral immunity of infant, we use a system serology approach to comprehensively profile antibody responses in a cohort of children ages 6 months to 5 years who were vaccinated with the mRNA-1273 COVID-19 vaccine (25 μg). Responses are compared with vaccinated adults (100 μg), in addition to naturally infected toddlers and young children. Despite their lower vaccine dose, vaccinated toddlers elicit a functional antibody response as strong as adults, with higher antibody-dependent phagocytosis compared to adults, without report of side effects. Moreover, mRNA vaccination is associated with a higher IgG3-dependent humoral profile against SARS-CoV-2 compared to natural infection, supporting that mRNA vaccination is effective at eliciting a robust antibody response in toddlers and young children.
Collapse
Affiliation(s)
- Nadège Nziza
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yixiang Deng
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lianna Wood
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Boston Children's Hospital, Department of Pediatric Gastroenterology, Boston, MA, USA
| | - Navneet Dhanoa
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
| | | | - Tina Chen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Abigail S Kane
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Zoe Swank
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jameson P Davis
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Melina Demokritou
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
| | - Anagha P Chitnis
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Alessio Fasano
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA, USA
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA, USA
| | - Nitya Jain
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bruce H Horwitz
- Harvard Medical School, Boston, MA, USA
- Boston Children's Hospital, Department of Emergency Medicine, Boston, MA, USA
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - David R Walt
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Wayne G Shreffler
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lael M Yonker
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA.
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
77
|
Madrid J, Agarwal P, Müller-Peltzer K, Askani M, Benning L, Selig M, Diehl P, Kalbhenn J, Trummer G, Utzolino S, Wengenmayer T, Busch HJ, Stolz D, Rieg S, Panning M, Schlett CL, Bamberg F, Askani E. Vaccination protects against acute respiratory distress syndrome (ARDS) in hospitalized patients with COVID-19. Clin Exp Med 2024; 24:21. [PMID: 38280024 PMCID: PMC10822002 DOI: 10.1007/s10238-023-01293-w] [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: 11/20/2023] [Accepted: 12/28/2023] [Indexed: 01/29/2024]
Abstract
This study aimed to analyze the effect of COVID-19 vaccination on the occurrence of ARDS in hospitalized COVID-19 patients. The study population of this retrospective, single-center cohort study consisted of hospitalized COVID-19 patients with known vaccination status and chest computed tomography imaging between July 2021 and February 2022. The impact of vaccination on ARDS in COVID-19 patients was assessed through logistic regression adjusting for demographic differences and confounding factors with statistical differences determined using confidence intervals and effect sizes. A total of 167 patients (69% male, average age 58 years, 95% CI [55; 60], 42% fully vaccinated) were included in the data analysis. Vaccinated COVID-19 patients had a reduced relative risk (RR) of developing ARDS (RR: 0.40, 95% CI [0.21; 0.62]). Consequently, non-vaccinated hospitalized patients had a 2.5-fold higher probability of developing ARDS. This risk reduction persisted after adjusting for several confounding variables (RR: 0.64, 95% CI [0.29; 0.94]) in multivariate analysis. The protective effect of COVID-19 vaccination increased with ARDS severity (RR: 0.61, 95% CI [0.37; 0.92]). Particularly, patients under 60 years old were at risk for ARDS onset and seemed to benefit from COVID-19 vaccination (RR: 0.51, 95% CI [0.20; 0.90]). COVID-19 vaccination showed to reduce the risk of ARDS occurrence in hospitalized COVID-19 patients, with a particularly strong effect in patients under 60 years old and those with more severe ARDS.
Collapse
Affiliation(s)
- Julian Madrid
- Department of Cardiology, Pneumology, Angiology, Acute Geriatrics and Intensive Care, Ortenau Klinikum, Klostenstraße 19, 77933, Lahr/Schwarzwald, Germany.
| | - Prerana Agarwal
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Katharina Müller-Peltzer
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Marvin Askani
- Department of Protestant Theology, Faculty of Theology, University of Heidelberg, Heidelberg, Germany
| | - Leo Benning
- University Emergency Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Diehl
- Department of Cardiology, Pneumology, Angiology, Acute Geriatrics and Intensive Care, Ortenau Klinikum, Klostenstraße 19, 77933, Lahr/Schwarzwald, Germany
| | - Johannes Kalbhenn
- Department of Anesthesiology and Intensive Care Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Georg Trummer
- Department of Cardiovascular Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Utzolino
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Wengenmayer
- Interdisciplinary Medical Intensive Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Jörg Busch
- University Emergency Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daiana Stolz
- Clinic of Respiratory Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Siegbert Rieg
- Division of Infectious Diseases, Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcus Panning
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Esther Askani
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| |
Collapse
|
78
|
Kwon O, Lee H, Jung J, Son YS, Jeon S, Yoo WD, Son N, Jung KB, Choi E, Lee IC, Kwon HJ, Kim C, Lee MO, Cho HS, Kim DS, Son MY. Chemically-defined and scalable culture system for intestinal stem cells derived from human intestinal organoids. Nat Commun 2024; 15:799. [PMID: 38280855 PMCID: PMC10821882 DOI: 10.1038/s41467-024-45103-7] [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: 02/02/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
Three-dimensional human intestinal organoids (hIO) are widely used as a platform for biological and biomedical research. However, reproducibility and challenges for large-scale expansion limit their applicability. Here, we establish a human intestinal stem cell (ISC) culture method expanded under feeder-free and fully defined conditions through selective enrichment of ISC populations (ISC3D-hIO) within hIO derived from human pluripotent stem cells. The intrinsic self-organisation property of ISC3D-hIO, combined with air-liquid interface culture in a minimally defined medium, forces ISC3D-hIO to differentiate into the intestinal epithelium with cellular diversity, villus-like structure, and barrier integrity. Notably, ISC3D-hIO is an ideal cell source for gene editing to study ISC biology and transplantation for intestinal diseases. We demonstrate the intestinal epithelium differentiated from ISC3D-hIO as a model system to study severe acute respiratory syndrome coronavirus 2 viral infection. ISC3D-hIO culture technology provides a biological tool for use in regenerative medicine and disease modelling.
Collapse
Affiliation(s)
- Ohman Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Hana Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Jaeeun Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Ye Seul Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Sojeong Jeon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Won Dong Yoo
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Naeun Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Kwang Bo Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Eunho Choi
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - In-Chul Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Republic of Korea
- KRIBB, Korea Preclinical Evaluation Center, Jeongeup, 56212, Republic of Korea
| | - Hyung-Jun Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Republic of Korea
- KRIBB, Korea Preclinical Evaluation Center, Jeongeup, 56212, Republic of Korea
| | - Chuna Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- KRIBB, Aging Convergence Research Center, Daejeon, 34141, Republic of Korea
| | - Mi-Ok Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dae Soo Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
79
|
Iakunchykova O, Leonardsen EH, Wang Y. Genetic evidence for causal effects of immune dysfunction in psychiatric disorders: where are we? Transl Psychiatry 2024; 14:63. [PMID: 38272880 PMCID: PMC10810856 DOI: 10.1038/s41398-024-02778-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
The question of whether immune dysfunction contributes to risk of psychiatric disorders has long been a subject of interest. To assert this hypothesis a plethora of correlative evidence has been accumulated from the past decades; however, a variety of technical and practical obstacles impeded on a cause-effect interpretation of these data. With the advent of large-scale omics technology and advanced statistical models, particularly Mendelian randomization, new studies testing this old hypothesis are accruing. Here we synthesize these new findings from genomics and genetic causal inference studies on the role of immune dysfunction in major psychiatric disorders and reconcile these new data with pre-omics findings. By reconciling these evidences, we aim to identify key gaps and propose directions for future studies in the field.
Collapse
Affiliation(s)
- Olena Iakunchykova
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Esten H Leonardsen
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway
| | - Yunpeng Wang
- Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, 0317, Oslo, Norway.
| |
Collapse
|
80
|
Vore AS, Marsland P, Barney TM, Varlinskaya EI, Landin JD, Healey KL, Kibble S, Swartzwelder HS, Chandler LJ, Deak T. Adolescent intermittent ethanol (AIE) produces lasting, sex-specific changes in rat body fat independent of changes in white blood cell composition. Front Physiol 2024; 15:1285376. [PMID: 38332987 PMCID: PMC10851431 DOI: 10.3389/fphys.2024.1285376] [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: 08/29/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Early initiation of alcohol use during adolescence, and adolescent binge drinking are risk factors for the development of alcohol use disorder later in life. Adolescence is a time of rapid sex-dependent neural, physiological, and behavioral changes as well as a period of heightened vulnerability to many effects of alcohol. The goal of the present studies was to determine age-related changes in blood (leukocyte populations) and body composition across adolescence and early adulthood, and to investigate whether adolescent intermittent ethanol (AIE) exposure would alter the trajectory of adolescent development on these broad physiological parameters. We observed significant ontogenetic changes in leukocyte populations that were mirrored by an age-related increase in cytokine expression among mixed populations of circulating leukocytes. Despite these developmental changes, AIE did not significantly alter overall leukocyte numbers or cytokine gene expression. However, AIE led to sex-specific changes in body fat mass and fat percentage, with AIE-exposed male rats showing significantly decreased fat levels and female rats showing significantly increased fat levels relative to controls. These changes suggest that while AIE may not alter overall leukocyte levels, more complex phenotypic changes in leukocyte populations could underlie previously reported differences in cytokine expression. Coupled with long-term shifts in adipocyte levels, this could have long-lasting effects on innate immunity and the capacity of individuals to respond to later immunological and physiological threats.
Collapse
Affiliation(s)
- Andrew S. Vore
- Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| | - Thaddeus M. Barney
- Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| | - Elena I. Varlinskaya
- Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| | - Justine D. Landin
- Department of Neurosciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States
| | - Kati L. Healey
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - Sandra Kibble
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - H. S. Swartzwelder
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
| | - Lawrence J. Chandler
- Department of Neurosciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, United States
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
81
|
Perusko M, Grundström J, Eldh M, Hamsten C, Apostolovic D, van Hage M. The α-Gal epitope - the cause of a global allergic disease. Front Immunol 2024; 15:1335911. [PMID: 38318181 PMCID: PMC10838981 DOI: 10.3389/fimmu.2024.1335911] [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: 11/09/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
The galactose-α-1,3-galactose (α-Gal) epitope is the cause of a global allergic disease, the α-Gal syndrome (AGS). It is a severe form of allergy to food and products of mammalian origin where IgE against the mammalian carbohydrate, α-Gal, is the cause of the allergic reactions. Allergic reactions triggered by parenterally administered α-Gal sources appear immediately, but those triggered via the oral route appear with a latency of several hours. The α-Gal epitope is highly immunogenic to humans, apes and old-world monkeys, all of which produce anti-α-Gal antibodies of the IgM, IgA and IgG subclasses. Strong evidence suggests that in susceptible individuals, class switch to IgE occurs after several tick bites. In this review, we discuss the strong immunogenic role of the α-Gal epitope and its structural resemblance to the blood type B antigen. We emphasize the broad abundance of α-Gal in different foods and pharmaceuticals and the allergenicity of various α-Gal containing molecules. We give an overview of the association of tick bites with the development of AGS and describe innate and adaptive immune response to tick saliva that possibly leads to sensitization to α-Gal. We further discuss a currently favored hypothesis explaining the mechanisms of the delayed effector phase of the allergic reaction to α-Gal. We highlight AGS from a clinical point of view. We review the different clinical manifestations of the disease and the prevalence of sensitization to α-Gal and AGS. The usefulness of various diagnostic tests is discussed. Finally, we provide different aspects of the management of AGS. With climate change and global warming, the tick density is increasing, and their geographic range is expanding. Thus, more people will be affected by AGS which requires more knowledge of the disease.
Collapse
Affiliation(s)
- Marija Perusko
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Innovative Centre of the Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Jeanette Grundström
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eldh
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carl Hamsten
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Danijela Apostolovic
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
82
|
Zawadzka A, Brzozowska B, Matyjanka A, Mikula M, Reszczyńska J, Tartas A, Fornalski KW. The Risk Function of Breast and Ovarian Cancers in the Avrami-Dobrzyński Cellular Phase-Transition Model. Int J Mol Sci 2024; 25:1352. [PMID: 38279352 PMCID: PMC10816518 DOI: 10.3390/ijms25021352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Specifying the role of genetic mutations in cancer development is crucial for effective screening or targeted treatments for people with hereditary cancer predispositions. Our goal here is to find the relationship between a number of cancerogenic mutations and the probability of cancer induction over the lifetime of cancer patients. We believe that the Avrami-Dobrzyński biophysical model can be used to describe this mechanism. Therefore, clinical data from breast and ovarian cancer patients were used to validate this model of cancer induction, which is based on a purely physical concept of the phase-transition process with an analogy to the neoplastic transformation. The obtained values of model parameters established using clinical data confirm the hypothesis that the carcinogenic process strongly follows fractal dynamics. We found that the model's theoretical prediction and population clinical data slightly differed for patients with the age below 30 years old, and that might point to the existence of an ancillary protection mechanism against cancer development. Additionally, we reveal that the existing clinical data predict breast or ovarian cancers onset two years earlier for patients with BRCA1/2 mutations.
Collapse
Affiliation(s)
- Anna Zawadzka
- Maria Skłodowska-Curie National Research Institute of Oncology (NIO-MSCI), 02-781 Warsaw, Poland; (A.Z.)
| | - Beata Brzozowska
- Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland; (B.B.)
| | - Anna Matyjanka
- Faculty of Physics, Warsaw University of Technology, 00-662 Warsaw, Poland
| | - Michał Mikula
- Maria Skłodowska-Curie National Research Institute of Oncology (NIO-MSCI), 02-781 Warsaw, Poland; (A.Z.)
| | - Joanna Reszczyńska
- Mossakowski Medical Research Institute, Polish Academy of Sciences (IMDiK PAN), 02-106 Warsaw, Poland;
| | - Adrianna Tartas
- Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland; (B.B.)
| | - Krzysztof W. Fornalski
- Faculty of Physics, Warsaw University of Technology, 00-662 Warsaw, Poland
- National Centre for Nuclear Research (NCBJ), 05-400 Otwock-Świerk, Poland
| |
Collapse
|
83
|
Benede N, Tincho MB, Walters A, Subbiah V, Ngomti A, Baguma R, Butters C, Hahnle L, Mennen M, Skelem S, Adriaanse M, Facey-Thomas H, Scott C, Day J, Spracklen TF, van Graan S, Balla SR, Moyo-Gwete T, Moore PL, MacGinty R, Botha M, Workman L, Johnson M, Goldblatt D, Zar HJ, Ntusi NA, Zühlke L, Webb K, Riou C, Burgers WA, Keeton RS. Distinct T cell polyfunctional profile in SARS-CoV-2 seronegative children associated with endemic human coronavirus cross-reactivity. iScience 2024; 27:108728. [PMID: 38235336 PMCID: PMC10792240 DOI: 10.1016/j.isci.2023.108728] [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: 07/10/2023] [Revised: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
SARS-CoV-2 infection in children typically results in asymptomatic or mild disease. There is a paucity of studies on SARS-CoV-2 antiviral immunity in African children. We investigated SARS-CoV-2-specific T cell responses in 71 unvaccinated asymptomatic South African children who were seropositive or seronegative for SARS-CoV-2. SARS-CoV-2-specific CD4+ T cell responses were detectable in 83% of seropositive and 60% of seronegative children. Although the magnitude of the CD4+ T cell response did not differ significantly between the two groups, their functional profiles were distinct, with SARS-CoV-2 seropositive children exhibiting a higher proportion of polyfunctional T cells compared to their seronegative counterparts. The frequency of SARS-CoV-2-specific CD4+ T cells in seronegative children was associated with the endemic human coronavirus (HCoV) HKU1 IgG response. Overall, the presence of SARS-CoV-2-responding T cells in seronegative children may result from cross-reactivity to endemic coronaviruses and could contribute to the relative protection from disease observed in SARS-CoV-2-infected children.
Collapse
Affiliation(s)
- Ntombi Benede
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Marius B. Tincho
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Avril Walters
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Vennesa Subbiah
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Amkele Ngomti
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Richard Baguma
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Claire Butters
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Lina Hahnle
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Mathilda Mennen
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Sango Skelem
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Marguerite Adriaanse
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
| | - Heidi Facey-Thomas
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Jonathan Day
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
| | - Timothy F. Spracklen
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Strauss van Graan
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Sashkia R. Balla
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L. Moore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- SA MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Rae MacGinty
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Maresa Botha
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Lesley Workman
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Heather J. Zar
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Medical Research Council (MRC) Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Ntobeko A.B. Ntusi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Liesl Zühlke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
- South African Medical Research Council, Francie Van Zijl Drive, Parow Cape Town, South Africa
| | - Kate Webb
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Observatory, South Africa
- Crick African Network, The Francis Crick Institute, London, UK
| | - Catherine Riou
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Wendy A. Burgers
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Observatory, South Africa
| | - Roanne S. Keeton
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Observatory, South Africa
| |
Collapse
|
84
|
Woo YR, Kim HS. Interaction between the microbiota and the skin barrier in aging skin: a comprehensive review. Front Physiol 2024; 15:1322205. [PMID: 38312314 PMCID: PMC10834687 DOI: 10.3389/fphys.2024.1322205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
The interplay between the microbes and the skin barrier holds pivotal significance in skin health and aging. The skin and gut, both of which are critical immune and neuroendocrine system, harbor microbes that are kept in balance. Microbial shifts are seen with aging and may accelerate age-related skin changes. This comprehensive review investigates the intricate connection between microbe dynamics, skin barrier, and the aging process. The gut microbe plays essential roles in the human body, safeguarding the host, modulating metabolism, and shaping immunity. Aging can perturb the gut microbiome which in turn accentuates inflammaging by further promoting senescent cell accumulation and compromising the host's immune response. Skin microbiota diligently upholds the epidermal barrier, adeptly fending off pathogens. The aging skin encompasses alterations in the stratum corneum structure and lipid content, which negatively impact the skin's barrier function with decreased moisture retention and increased vulnerability to infection. Efficacious restoration of the skin barrier and dysbiosis with strategic integration of acidic cleansers, emollients with optimal lipid composition, antioxidants, and judicious photoprotection may be a proactive approach to aging. Furthermore, modulation of the gut-skin axis through probiotics, prebiotics, and postbiotics emerges as a promising avenue to enhance skin health as studies have substantiated their efficacy in enhancing hydration, reducing wrinkles, and fortifying barrier integrity. In summary, the intricate interplay between microbes and skin barrier function is intrinsically woven into the tapestry of aging. Sound understanding of these interactions, coupled with strategic interventions aimed at recalibrating the microbiota and barrier equilibrium, holds the potential to ameliorate skin aging. Further in-depth studies are necessary to better understand skin-aging and develop targeted strategies for successful aging.
Collapse
Affiliation(s)
- Yu Ri Woo
- Department of Dermatology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hei Sung Kim
- Department of Dermatology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
85
|
Beverley J, Babcock S, Carvalho G, Cowell LG, Duesing S, He Y, Hurley R, Merrell E, Scheuermann RH, Smith B. Coordinating virus research: The Virus Infectious Disease Ontology. PLoS One 2024; 19:e0285093. [PMID: 38236918 PMCID: PMC10796065 DOI: 10.1371/journal.pone.0285093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/12/2023] [Indexed: 01/22/2024] Open
Abstract
The COVID-19 pandemic prompted immense work on the investigation of the SARS-CoV-2 virus. Rapid, accurate, and consistent interpretation of generated data is thereby of fundamental concern. Ontologies-structured, controlled, vocabularies-are designed to support consistency of interpretation, and thereby to prevent the development of data silos. This paper describes how ontologies are serving this purpose in the COVID-19 research domain, by following principles of the Open Biological and Biomedical Ontology (OBO) Foundry and by reusing existing ontologies such as the Infectious Disease Ontology (IDO) Core, which provides terminological content common to investigations of all infectious diseases. We report here on the development of an IDO extension, the Virus Infectious Disease Ontology (VIDO), a reference ontology covering viral infectious diseases. We motivate term and definition choices, showcase reuse of terms from existing OBO ontologies, illustrate how ontological decisions were motivated by relevant life science research, and connect VIDO to the Coronavirus Infectious Disease Ontology (CIDO). We next use terms from these ontologies to annotate selections from life science research on SARS-CoV-2, highlighting how ontologies employing a common upper-level vocabulary may be seamlessly interwoven. Finally, we outline future work, including bacteria and fungus infectious disease reference ontologies currently under development, then cite uses of VIDO and CIDO in host-pathogen data analytics, electronic health record annotation, and ontology conflict-resolution projects.
Collapse
Affiliation(s)
- John Beverley
- Department of Philosophy, University at Buffalo, Buffalo, NY, United States of America
- National Center for Ontological Research, Buffalo, NY, United States of America
| | - Shane Babcock
- National Center for Ontological Research, Buffalo, NY, United States of America
- Air Force Research Laboratory, Wright Patterson Air Force Base, Riverside, OH, United States of America
| | - Gustavo Carvalho
- Department of Cognitive Science, Northwestern University, Evanston, IL, United States of America
| | - Lindsay G. Cowell
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Sebastian Duesing
- Department of Philosophy, Loyola University, Chicago, IL, United States of America
| | - Yongqun He
- Computational Medicine and Bioinformatics, University of Michigan Medical School, He Group, Ann Arbor, MI, United States of America
| | - Regina Hurley
- National Center for Ontological Research, Buffalo, NY, United States of America
- Department of Philosophy, Northwestern University, Evanston, IL, United States of America
| | - Eric Merrell
- Department of Philosophy, University at Buffalo, Buffalo, NY, United States of America
- National Center for Ontological Research, Buffalo, NY, United States of America
| | - Richard H. Scheuermann
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA, United States of America
- Department of Pathology, University of California, San Diego, CA, United States of America
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States of America
| | - Barry Smith
- Department of Philosophy, University at Buffalo, Buffalo, NY, United States of America
- National Center for Ontological Research, Buffalo, NY, United States of America
| |
Collapse
|
86
|
Hong E, Mao J, Ke Z, Tao W. Knowledge, attitudes and practices towards community-acquired pneumonia and COVID-19 among general population: a cross-sectional study. Antimicrob Resist Infect Control 2024; 13:6. [PMID: 38233911 PMCID: PMC10795257 DOI: 10.1186/s13756-023-01361-6] [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: 07/20/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND This study aimed to assess the knowledge, attitudes, and practices (KAP) of the general population to community-acquired pneumonia (CAP) and COVID-19. METHODS A cross-sectional study was conducted between September 2022 and February 2023, involving the general population from Ningbo Municipal Hospital of Traditional Chinese Medicine with a self-developed questionnaire. RESULTS A total of 637 valid questionnaires were collected, with the majority of participants being female (62.48%). The mean score for knowledge, attitudes, and practices were 7.60 ± 2.39 (possible range: 0-12), 43.20 ± 4.57 (possible range: 11-55), and 34.57 ± 4.95 (possible range: 10-50), respectively. Multivariate logistic regression analysis indicated that master's degree or above (OR = 6.04, 95% CI: 1.80-20.31, P = 0.004) and occupation in business or service careers (OR = 0.28, 95% CI: 0.17-0.48, P < 0.001) were independent associated with knowledge. The knowledge (OR = 1.32, 95%CI: 1.20-1.44, P < 0.001) and female gender (OR = 1.48, 95%CI: 1.03-2.14, P = 0.036) were independently associated with positive attitudes. Attitudes (OR = 1.34, 95%CI: 1.26-1.43, P < 0.001) and a monthly household income greater than 20,000 RMB (OR = 0.31, 95%CI: 0.15-0.64, P = 0.001) were independent associated with practices. Pearson correlation analysis revealed that knowledge positively correlated with attitude scores (r = 0.348, P < 0.001) and practice scores (r = 0.259, P < 0.001), and attitude and practice scores were also positively correlated (r = 0.563, P < 0.001). Structural equation modeling showed that knowledge predicted attitudes (β = 0.67, P < 0.001) and practices (β = 0.17, P = 0.017), while attitudes predicted practices (β = 0.58, P < 0.001). CONCLUSION General population had moderate knowledge, positive attitudes and average practices towards CAP and COVID-19.
Collapse
Affiliation(s)
- Er Hong
- Department of Respiratory, Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo, 315010, China.
| | - Jia Mao
- Department of Respiratory, Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo, 315010, China
| | - Zhicheng Ke
- Department of Respiratory, Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo, 315010, China
| | - Wei Tao
- Department of Radiology, Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo, 315010, China
| |
Collapse
|
87
|
de Graaf SC, Bondt A, van Rijswijck DMH, Juncker HG, Mulleners SJ, Damen MJA, Hoek M, van Keulen BJ, van Goudoever JB, Heck AJR, Dingess KA. A case series exploring the human milk polyclonal IgA1 response to repeated SARS-CoV-2 vaccinations by LC-MS based fab profiling. Front Nutr 2024; 10:1305086. [PMID: 38288064 PMCID: PMC10822949 DOI: 10.3389/fnut.2023.1305086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Upon vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) humans will start to produce antibodies targeting virus specific antigens that will end up in circulation. In lactating women such antibodies will also end up in breastmilk, primarily in the form of secretory immunoglobulin A1 (SIgA1), the most abundant immunoglobulin (Ig) in human milk. Here we set out to investigate the SIgA1 clonal repertoire response to repeated SARS-CoV-2 vaccination, using a LC-MS fragment antigen-binding (Fab) clonal profiling approach. Methods We analyzed the breastmilk of six donors from a larger cohort of 109 lactating mothers who received one of three commonly used SARS-CoV-2 vaccines. We quantitatively monitored the SIgA1 Fab clonal profile over 16 timepoints, from just prior to the first vaccination until 15 days after the second vaccination. Results In all donors, we detected a population of 89-191 vaccine induced clones. These populations were unique to each donor and heterogeneous with respect to individual clonal concentrations, total clonal titer, and population size. The vaccine induced clones were dominated by persistent clones (68%) which came up after the first vaccination and were retained or reoccurred after the second vaccination. However, we also observe transient SIgA1 clones (16%) which dissipated before the second vaccination, and vaccine induced clones which uniquely emerged only after the second vaccination (16%). These distinct populations were observed in all analyzed donors, regardless of the administered vaccine. Discussion Our findings suggest that while individual donors have highly unique human milk SIgA1 clonal profiles and a highly personalized SIgA1 response to SARS-CoV-2 vaccination, there are also commonalities in vaccine induced responses.
Collapse
Affiliation(s)
- Sebastiaan C. de Graaf
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Albert Bondt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Danique M. H. van Rijswijck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Hannah G. Juncker
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Sien J. Mulleners
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mirjam J. A. Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Max Hoek
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Britt J. van Keulen
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Johannes B. van Goudoever
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
| | - Kelly A. Dingess
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, Netherlands
- Netherlands Proteomics Center, Utrecht, Netherlands
- Department of Pediatrics, Amsterdam Reproduction and Development Research Institute, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
88
|
Giugni FR, Duarte-Neto AN, da Silva LFF, Monteiro RAA, Mauad T, Saldiva PHN, Dolhnikoff M. Younger age is associated with cardiovascular pathological phenotype of severe COVID-19 at autopsy. Front Med (Lausanne) 2024; 10:1327415. [PMID: 38259848 PMCID: PMC10801169 DOI: 10.3389/fmed.2023.1327415] [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: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction COVID-19 affects patients of all ages. There are few autopsy studies focusing on the younger population. We assessed an autopsy cohort aiming to understand how age influences pathological outcomes in fatal COVID-19. Methods This study included autopsied patients, aged 6 months to 83 years, with confirmed COVID-19 in 2020-2021. We collected tissue samples from deceased patients using a minimally invasive autopsy protocol and assessed pathological data following a systematic approach. Results Eighty-six patients were included, with a median age of 55 years (IQR 32.3-66.0). We showed that age was significantly lower in patients with acute heart ischemia (p = 0.004), myocarditis (p = 0.03) and lung angiomatosis (p < 0.001), and significantly higher in patients with exudative diffuse alveolar damage (p = 0.02), proliferative diffuse alveolar damage (p < 0.001), lung squamous metaplasia (p = 0.003) and lung viral atypia (p = 0.03), compared to patients without those findings. We stratified patients by their age and showed that cardiovascular findings were more prevalent in children and young adults. We performed principal component analysis and cluster of pathological variables, and showed that cardiovascular variables clustered and covariated together, and separated from pulmonary variables. Conclusion We showed that age modulates pathological outcomes in fatal COVID-19. Younger age is associated with cardiovascular abnormalities and older age with pulmonary findings.
Collapse
Affiliation(s)
- Fernando R. Giugni
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Amaro N. Duarte-Neto
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Fernando F. da Silva
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- SVOC - Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| | - Renata A. A. Monteiro
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Thais Mauad
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo H. N. Saldiva
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- LIM 05 - Laboratório de Patologia Ambiental e Experimental, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
89
|
Leonard J, Ladner L, Harris EA, de Jager C, Theus MH. The Neuroimmune Interface: Age-Related Responses to Traumatic Brain Injury. ADVANCES IN NEUROBIOLOGY 2024; 42:241-262. [PMID: 39432046 DOI: 10.1007/978-3-031-69832-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Traumatic Brain Injury (TBI) is a significant public health issue, with diverse consequences across the lifespan. This comprehensive review explores the complex interplay between age-related responses and the immune system following TBI. TBI exhibits distinct effects in pediatric, adult, and elderly populations, with profound implications for recovery and long-term outcomes. The immune system, as a key player in the post-TBI inflammatory cascade, exerts age-dependent influences on inflammation, neuroinflammation, and tissue repair. We examine the evolving understanding of age-related neuroinflammatory responses, cytokine profiles, and the role of immune cells, such as microglia and T cells, in the context of TBI. Furthermore, we evaluate the therapeutic implications of age-specific immunomodulation strategies toward mitigating TBI-associated neuropathology. This review consolidates the current knowledge on age-related immune responses in TBI, shedding light on potential avenues for tailored therapeutic interventions across the age spectrum. Understanding these nuanced responses is crucial for optimizing patient care and enhancing recovery outcomes in the aftermath of traumatic brain injury.
Collapse
Affiliation(s)
- John Leonard
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Liliana Ladner
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Elizabeth A Harris
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Caroline de Jager
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Michelle H Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
90
|
Lobova OV, Avramenko IV, Shpak II. COVID-19 associated anosmia in pediatric patients: subject publications review. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:114-119. [PMID: 38431815 DOI: 10.36740/wlek202401114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To review the publications subject to the problem of COVID-19 associated anosmia incidence in pediatric patients as well as its pathogenesis, diagnostics, treatment and recovery. The peculiarity of pediatric COVID-19 anosmia is due to children accounting for very low percentage of COVID-19 patients (comparing to one in adults), mostly with milder course of the disease. Awareness of anosmia and its proper diagnostics is crucial in children and adolescents, considering it can be the only manifestation in COVID-19 positive pediatric patients. PATIENTS AND METHODS Materials and Methods: In order to achieve this goal a meta-analysis of information from databases followed by statistical processing and generalisation of the obtained data was carried out. CONCLUSION Conclusions: Publications on COVID-19 anosmia in children and adolescents are less numerous than those concerning adult patients, so it is important to use every single trustworthy one. Anosmia/ageusia may be the only symptom, early identifier and the strongest predictor of COVID-19 infection in pediatric patients. Prospects for further scientific researches. Further researches regarding differential diagnostics of COVID-19 and other infections, including seasonal influenza, manifesting with both olfactory and taste dysfunction as well as anosmia diagnostics in children and adolescents with autistic spectrum and different types of mental disorders are possible.
Collapse
Affiliation(s)
| | - Iryna V Avramenko
- KYIV MEDICAL UNIVERSITY, KYIV, UKRAINE; DNIPRO STATE MEDICAL UNIVERSITY, DNIPRO, UKRAINE
| | | |
Collapse
|
91
|
Chitre SD, Crews CM, Tessema MT, Plėštytė-Būtienė I, Coffee M, Richardson ET. The impact of anthropogenic climate change on pediatric viral diseases. Pediatr Res 2024; 95:496-507. [PMID: 38057578 PMCID: PMC10872406 DOI: 10.1038/s41390-023-02929-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/12/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
The adverse effects of climate change on human health are unfolding in real time. Environmental fragmentation is amplifying spillover of viruses from wildlife to humans. Increasing temperatures are expanding mosquito and tick habitats, introducing vector-borne viruses into immunologically susceptible populations. More frequent flooding is spreading water-borne viral pathogens, while prolonged droughts reduce regional capacity to prevent and respond to disease outbreaks with adequate water, sanitation, and hygiene resources. Worsening air quality and altered transmission seasons due to an increasingly volatile climate may exacerbate the impacts of respiratory viruses. Furthermore, both extreme weather events and long-term climate variation are causing the destruction of health systems and large-scale migrations, reshaping health care delivery in the face of an evolving global burden of viral disease. Because of their immunological immaturity, differences in physiology (e.g., size), dependence on caregivers, and behavioral traits, children are particularly vulnerable to climate change. This investigation into the unique pediatric viral threats posed by an increasingly inhospitable world elucidates potential avenues of targeted programming and uncovers future research questions to effect equitable, actionable change. IMPACT: A review of the effects of climate change on viral threats to pediatric health, including zoonotic, vector-borne, water-borne, and respiratory viruses, as well as distal threats related to climate-induced migration and health systems. A unique focus on viruses offers a more in-depth look at the effect of climate change on vector competence, viral particle survival, co-morbidities, and host behavior. An examination of children as a particularly vulnerable population provokes programming tailored to their unique set of vulnerabilities and encourages reflection on equitable climate adaptation frameworks.
Collapse
Affiliation(s)
- Smit D Chitre
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Cecilia M Crews
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Mesfin Teklu Tessema
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA.
- International Rescue Committee, New York, NY, USA.
| | | | - Megan Coffee
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
- International Rescue Committee, New York, NY, USA
- New York University Grossman School of Medicine, New York, NY, USA
| | - Eugene T Richardson
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
92
|
Alladin SNB, Berry D, Anisimova E, Judson R, Whittaker P, Dalmaijer ES. Children aged 5-13 years show adult-like disgust avoidance, but not proto-nausea. Brain Neurosci Adv 2024; 8:23982128241279616. [PMID: 39247223 PMCID: PMC11380130 DOI: 10.1177/23982128241279616] [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: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Disgust is a vital emotion in the avoidance of illness. Human adults across cultures show disgust towards sources of potential contamination or pathogens, and elect to avoid their ingestion or even to look at them. Stomach rhythms appear to play an important role: disgust reduces normogastric power, and the pharmacological normalisation of gastric state reduces disgust avoidance. Human children are remarkably slow to develop disgust as measured by self-report and facial expressions. Here, we investigate whether disgust-induced avoidance (measured using eye tracking) and changes in gastric rhythm (measured using electrogastrography) exist in children aged 5 to 13 years (N = 45). We found that children in this bracket showed oculomotor avoidance of disgusting stimuli in a preferential-looking task, similar to adult samples in previous research. However, in contrast to adult samples in previous research, children did not show an attenuation in normogastric power. These findings could suggest that avoidance behaviour precedes gastric involvement during disgust. This would support the idea that children initially respond to parental modelling: parents set (and enforce) the social norm of disgust avoidance, and children initially conform and only later do they internalise disgust as an interoceptive signal. Alternatively, the employed stimuli could have been potent enough to induce oculomotor avoidance, but not a gastric response. Research is slim in this area, and future work should focus on elucidating the role of the stomach in disgust, and on longitudinal studies of disgust development from childhood to adolescence.
Collapse
Affiliation(s)
| | - Dani Berry
- School of Psychological Science, University of Bristol, Bristol, UK
| | | | - Ruth Judson
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Poppy Whittaker
- School of Psychological Science, University of Bristol, Bristol, UK
| | | |
Collapse
|
93
|
Biliute G, Miskinyte M, Miskiniene A, Zinkeviciene A, Kvedariene V. Sensitization profiles to house dust mite Dermatophagoides pteronyssinus molecular allergens in the Lithuanian population: Understanding allergic sensitization patterns. Clin Transl Allergy 2024; 14:e12332. [PMID: 38282198 PMCID: PMC10807355 DOI: 10.1002/clt2.12332] [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: 09/27/2023] [Revised: 12/11/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND House dust mite (HDM) allergy is a prevalent global health concern, with varying sensitization profiles observed across populations. We aimed to provide a comprehensive assessment of molecular allergen sensitization patterns in the Lithuanian population, with a focus on Dermatophagoides pteronyssinus (Der p), and investigate patterns of concomitant reactivity among different allergens to enhance the accuracy of HDM allergy diagnostics. METHODS A comprehensive analysis of 1520 patient test results in Lithuania from 2020 to 2022 was performed. Sensitization patterns to major (Der p 1, Der p 2, and Der p 23) and minor (Der p 5, Der p 7, and Der p 21) Der p allergen components were described using molecular-based diagnostics. Additionally, we investigated sensitization to allergen components from other allergen sources, including tropomyosins (Der p 10, Per a 7, Pen m 1, Ani s 3, Blo t 10) and arginine kinases (Pen m 2, Bla g 9, Der p 20). RESULTS This study reveals a high prevalence of HDM sensitization in Lithuania - 481 individuals (45.38% of the sensitized group) exhibited sensitization to at least one Der p allergen component. Importantly, within the sensitized group, 37.21% of patients were sensitized to Der p 5, Der p 7, or Der p 21 in addition to major allergenic components. Distinct sensitization patterns were observed across different age groups, indicating the influence of age-related factors. Furthermore, we confirmed cross-reactivity between Der p 5 and Blo t 5 as well as between Der p 21 and Blo t 21, emphasizing the clinical relevance of these associations. We also highlighted the complexity of sensitization patterns among tropomyosins and arginine kinases. CONCLUSION This study provides valuable insights into HDM allergy sensitization profiles in Lithuania, emphasizing the importance of considering major and minor HDM allergen components for accurate diagnosis and management of HDM-related allergic diseases. Differences between populations and age-related factors impact sensitization patterns. Understanding concomitant reactivity among allergens, such as Der p 5 and Blo t 5, Der p 21 and Blo t 21, tropomyosins, and arginine kinases, is crucial for improving diagnostic strategies and developing targeted interventions for allergic individuals.
Collapse
Affiliation(s)
- Gabija Biliute
- Faculty of MedicineClinic of Chest DiseasesAllergology and ImmunologyInstitute of Clinical MedicineVilnius UniversityVilniusLithuania
| | | | | | - Aukse Zinkeviciene
- State Research Institute Centre for Innovative MedicineDepartment of ImmunologyVilnius UniversityVilniusLithuania
| | - Violeta Kvedariene
- Faculty of MedicineClinic of Chest DiseasesAllergology and ImmunologyInstitute of Clinical MedicineVilnius UniversityVilniusLithuania
- Department of PathologyFaculty of MedicineInstitute of Biomedical SciencesVilnius UniversityVilniusLithuania
| |
Collapse
|
94
|
Clarkson KA, Porter CK, Talaat KR, Kapulu MC, Chen WH, Frenck RW, Bourgeois AL, Kaminski RW, Martin LB. Shigella-Controlled Human Infection Models: Current and Future Perspectives. Curr Top Microbiol Immunol 2024; 445:257-313. [PMID: 35616717 PMCID: PMC7616482 DOI: 10.1007/82_2021_248] [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: 10/18/2022]
Abstract
Shigella-controlled human infection models (CHIMs) are an invaluable tool utilized by the vaccine community to combat one of the leading global causes of infectious diarrhea, which affects infants, children and adults regardless of socioeconomic status. The impact of shigellosis disproportionately affects children in low- and middle-income countries (LMICs) resulting in cognitive and physical stunting, perpetuating a cycle that must be halted. Shigella-CHIMs not only facilitate the early evaluation of enteric countermeasures and up-selection of the most promising products but also provide insight into mechanisms of infection and immunity that are not possible utilizing animal models or in vitro systems. The greater understanding of shigellosis obtained in CHIMs builds and empowers the development of new generation solutions to global health issues which are unattainable in the conventional laboratory and clinical settings. Therefore, refining, mining and expansion of safe and reproducible infection models hold the potential to create effective means to end diarrheal disease and associated co-morbidities associated with Shigella infection.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway Street Hampton House, Baltimore, MD, 21205, USA
| | - Melissa C Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi County Hospital, Off Bofa Road, Kilifi, 80108, Kenya
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - A Louis Bourgeois
- PATH Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Washington, DC, 20001, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
95
|
Le Bras C, Rault L, Jacquet N, Daniel N, Chuat V, Valence F, Bellanger A, Bousarghin L, Blat S, Le Loir Y, Le Huërou-Luron I, Even S. Two human milk-like synthetic bacterial communities displayed contrasted impacts on barrier and immune responses in an intestinal quadricellular model. ISME COMMUNICATIONS 2024; 4:ycad019. [PMID: 38415201 PMCID: PMC10897888 DOI: 10.1093/ismeco/ycad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 02/29/2024]
Abstract
The human milk (HM) microbiota, a highly diverse microbial ecosystem, is thought to contribute to the health benefits associated with breast-feeding, notably through its impact on infant gut microbiota. Our objective was to further explore the role of HM bacteria on gut homeostasis through a "disassembly/reassembly" strategy. HM strains covering the diversity of HM cultivable microbiota were first characterized individually and then assembled in synthetic bacterial communities (SynComs) using two human cellular models, peripheral blood mononuclear cells and a quadricellular model mimicking intestinal epithelium. Selected HM bacteria displayed a large range of immunomodulatory properties and had variable effects on epithelial barrier, allowing their classification in functional groups. This multispecies characterization of HM bacteria showed no clear association between taxonomy and HM bacteria impacts on epithelial immune and barrier functions, revealing the entirety and complexity of HM bacteria potential. More importantly, the assembly of HM strains into two SynComs of similar taxonomic composition but with strains exhibiting distinct individual properties, resulted in contrasting impacts on the epithelium. These impacts of SynComs partially diverged from the predicted ones based on individual bacteria. Overall, our results indicate that the functional properties of the HM bacterial community rather than the taxonomic composition itself could play a crucial role in intestinal homeostasis of infants.
Collapse
Affiliation(s)
- Charles Le Bras
- STLO, INRAE, Institut Agro, Rennes, 35042, France
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Lucie Rault
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| | | | | | | | | | | | - Latifa Bousarghin
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Yves Le Loir
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| | | | - Sergine Even
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| |
Collapse
|
96
|
Miron VD, Raianu RO, Filimon C, Craiu M. Clinical Differences between SARS-CoV-2 and RSV Infections in Infants: Findings from a Case-Control Study. Viruses 2023; 16:63. [PMID: 38257763 PMCID: PMC10819890 DOI: 10.3390/v16010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Infants are a unique pediatric group due to their high hospitalization rates and unfavorable outcomes from acute infectious diseases. Understanding the clinical differences and aftereffects of SARS-CoV-2 in comparison to other prevalent viruses in this age group, like RSV, is crucial for effective management. We conducted a retrospective case-control study of infants hospitalized with SARS-CoV-2 or respiratory syncytial virus (RSV) infection in one year, in a tertiary pediatric hospital in Bucharest, Romania. A total of 188 infants were included in the analysis in a 1:1 ratio (94 with SARS-CoV-2 infection and 94 with RSV infection). Infants with COVID-19 were 10.2 times more likely to have fever (p < 0.001) and 2.4 times more likely to have diarrhea (p = 0.016). Conversely, infants with RSV were 2.5 times more likely to have a cough (p < 0.001), 3.0 times more likely to have nasal congestion (p < 0.001), and 14.7 times more likely to present with dyspnea (p < 0.001). Increased lymphocyte count was more common in infants with RSV (p = 0.008), while lymphopenia was more frequent in infants with SARS-CoV-2 (p = 0.011). The median length of hospital stay was one day longer in infants with RSV infection (5 days vs. 4 days). Overall, infants with RSV infection had a 27.3-fold increased risk of developing respiratory failure (p < 0.001), while infants with COVID-19 had a 5.8-fold increased risk of laryngitis (p = 0.003). Our findings suggest that infants with SARS-CoV-2 infection may present with polymorphic symptoms, mostly dominated by fever, whereas infants with RSV often present with respiratory symptoms. Laboratory differentiation between the two infections is challenging; therefore, the use of rapid antigen or molecular diagnostic tests is crucial for accurate diagnosis, epidemiologically appropriate measures, and effective management. Continued surveillance of both viruses in infants, and beyond, and the implementation of specific control measures are needed to mitigate their impact on this vulnerable pediatric group.
Collapse
Affiliation(s)
| | - Raluca-Oana Raianu
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Claudiu Filimon
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihai Craiu
- Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 020395 Bucharest, Romania
| |
Collapse
|
97
|
Li X, Brejnrod A, Thorsen J, Zachariasen T, Trivedi U, Russel J, Vestergaard GA, Stokholm J, Rasmussen MA, Sørensen SJ. Differential responses of the gut microbiome and resistome to antibiotic exposures in infants and adults. Nat Commun 2023; 14:8526. [PMID: 38135681 PMCID: PMC10746713 DOI: 10.1038/s41467-023-44289-6] [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: 03/20/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Despite their crucial importance for human health, there is still relatively limited knowledge on how the gut resistome changes or responds to antibiotic treatment across ages, especially in the latter case. Here, we use fecal metagenomic data from 662 Danish infants and 217 young adults to fill this gap. The gut resistomes are characterized by a bimodal distribution driven by E. coli composition. The typical profile of the gut resistome differs significantly between adults and infants, with the latter distinguished by higher gene and plasmid abundances. However, the predominant antibiotic resistance genes (ARGs) are the same. Antibiotic treatment reduces bacterial diversity and increased ARG and plasmid abundances in both cohorts, especially core ARGs. The effects of antibiotic treatments on the gut microbiome last longer in adults than in infants, and different antibiotics are associated with distinct impacts. Overall, this study broadens our current understanding of gut resistome dynamics and the impact of antibiotic treatment across age groups.
Collapse
Affiliation(s)
- Xuanji Li
- Department of Biology, Section of Microbiology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Asker Brejnrod
- Department of Health Technology, Technical University of Denmark, Section of Bioinformatics, 2800 Kgs, Lyngby, Denmark
| | - Jonathan Thorsen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Trine Zachariasen
- Department of Health Technology, Technical University of Denmark, Section of Bioinformatics, 2800 Kgs, Lyngby, Denmark
| | - Urvish Trivedi
- Department of Biology, Section of Microbiology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Jakob Russel
- Department of Biology, Section of Microbiology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Gisle Alberg Vestergaard
- Department of Health Technology, Technical University of Denmark, Section of Bioinformatics, 2800 Kgs, Lyngby, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Food Science, Section of Microbiology and Fermentation, University of Copenhagen, 1958, Frederiksberg C, Denmark
| | - Morten Arendt Rasmussen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
- Department of Food Science, Section of Microbiology and Fermentation, University of Copenhagen, 1958, Frederiksberg C, Denmark.
| | - Søren Johannes Sørensen
- Department of Biology, Section of Microbiology, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
98
|
Zuurveld M, Diks MAP, Kiliaan PCJ, Garssen J, Folkerts G, van’t Land B, Willemsen LEM. Butyrate interacts with the effects of 2'FL and 3FL to modulate in vitro ovalbumin-induced immune activation, and 2'FL lowers mucosal mast cell activation in a preclinical model for hen's egg allergy. Front Nutr 2023; 10:1305833. [PMID: 38174112 PMCID: PMC10762782 DOI: 10.3389/fnut.2023.1305833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
Background Early life provides a window of opportunity to prevent allergic diseases. With a prevalence of 0.5-2% in infants, hen's egg allergy is one of the most common food allergies. The immunomodulatory effects of human milk oligosaccharides (HMOs), 2'-fucosyllactose (2'FL), and 3-fucosyllactose (3FL) were studied in an in vitro mucosal immune model and an in vivo murine model for hen's egg (ovalbumin) allergy. Methods Intestinal epithelial cell (IEC)/dendritic cell (DC) and DC/T cell cocultures were used to expose IECs to ovalbumin (OVA) in an in vitro mucosal immune model. The effects of epithelial pre-incubation with 0.1% 2'FL or 3FL and/or 0.5 mM butyrate were studied. Three- to four-weeks-old female C3H/HeOuJ mice were fed AIN93G diets containing 0.1-0.5% 2'FL or 3FL 2 weeks before and during OVA sensitization and challenge. Allergic symptoms and systemic and local immune parameters were assessed. Results Exposing IECs to butyrate in vitro left the IEC/DC/T cell cross-talk unaffected, while 2'FL and 3FL showed differential immunomodulatory effects. In 3FL exposed IEC-DC-T cells, the secretion of IFNγ and IL10 was enhanced. This was observed upon pre-incubation of IECs with 2'FL and butyrate as well, but not 2'FL alone. The presence of butyrate did not affect OVA activation, but when combined with 3FL, an increase in IL6 release from DCs was observed (p < 0.001). OVA allergic mice receiving 0.5% 3FL diet had a lower %Th2 cells in MLNs, but the humoral response was unaltered compared to control mice. OVA-allergic mice receiving 0.1 or 0.5% 2'FL diets had lower serum levels of OVA-IgG2a (p < 0.05) or the mast cell marker mMCP1, in association with increased concentration of cecal short-chain fatty acids (SCFAs) (p < 0.05). Conclusion In vitro butyrate exposure promotes the development of a downstream type 1 and regulatory response observed after 2'FL exposure. 2'FL and 3FL differentially modulate ovalbumin-induced mucosal inflammation predominantly independent of butyrate. Mice receiving dietary 3FL during ovalbumin sensitization and challenge had lowered Th2 activation while the frequency of Treg cells was enhanced. By contrast, 2'FL improved the humoral immune response and suppressed mast cell activation in association with increased SCFAs production in the murine model for hen's egg allergy.
Collapse
Affiliation(s)
- M. Zuurveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - M. A. P. Diks
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - P. C. J. Kiliaan
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - J. Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research B.V, Utrecht, Netherlands
| | - G. Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - B. van’t Land
- Danone Nutricia Research B.V, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - L. E. M. Willemsen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
99
|
Martins AF, Santos ASE, Moreira JC, Câmara VDM, Asmus CIRF, Rosa ACS, Vineis P, Meyer A. Exposure of pregnant women and their children to pyrethroid insecticides in Rio de Janeiro, Brazil. Front Public Health 2023; 11:1274724. [PMID: 38162602 PMCID: PMC10756647 DOI: 10.3389/fpubh.2023.1274724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Background Pyrethroids are commonly used insecticides in Brazil. Gestational and early childhood exposure to pyrethroids has been linked to adverse health effects, including neurodevelopmental delays, behavioral issues, and endocrine disruption. This study evaluated the exposure of pregnant women and their children to pyrethroid insecticides in Rio de Janeiro, Brazil. Methods Creatinine-adjusted levels of the pyrethroid metabolites 3-phenoxy benzoic acid (3-PBA) and 4-fluoro-3-phenoxybenzyl acid (4-FPBA) were measured in the urine of 142 pregnant women and their children at birth and in the first, third, and 6th months of life. Results The geometric mean (GM) and 95% confidence interval (95% CI) of 3-PBA and 4-FPBA urinary concentrations in pregnant women were 0.50 (0.37-0.67) and 0.37 (0.05-2.90) ng/mg, detected in 47.2 and 10.6%, respectively. Urinary concentrations of 3-PBA in the children were 0.18 (0.15-0.23) ng/mg at birth, 0.36 (0.08-1.56) ng/mg at 1-month-old, 0.68 (0.36-1.27) ng/mg at 3-month-old, and 1.36 (0.77-2.42) ng/mg at 6-month-old, and the detection rates were respectively 10.8, 9.4, 20.9, and 20.7%. Discussion This study is one of the few that has evaluated the urinary concentrations of pyrethroids in newborns and children in their 1st year of life. The results of this study show that children's exposure to pyrethroids significantly increases after birth.
Collapse
Affiliation(s)
- Amanda Friaes Martins
- Public Health Program, Public Health Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Souza Espindola Santos
- Occupational and Environmental Health Branch, Public Health Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Josino Costa Moreira
- Occupational and Environmental Health Branch, Public Health Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Volney de Magalhaes Câmara
- Occupational and Environmental Health Branch, Public Health Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Cristina Simoes Rosa
- Center for Studies of Human Ecology and Worker's Health, National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Paolo Vineis
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Armando Meyer
- Occupational and Environmental Health Branch, Public Health Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
100
|
Maleki A, Mehrbod P, Bokharaei-Salim F, Eybpoosh S, Tavakoli M, Mohammadnejad AE, Hosseini Z, Kashanian S, Asadi LF, Salehi-Vaziri M, Fotouhi F. Epidemiological surveillance of respiratory viral infections in SARS-CoV-2-negative samples during COVID-19 pandemic in Iran. Virol J 2023; 20:296. [PMID: 38093303 PMCID: PMC10720196 DOI: 10.1186/s12985-023-02226-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/02/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND To improve the patient care, public health surveillance, and infection control, it is crucial to identify the presence and frequency of the common respiratory infections in individuals with COVID-19 symptoms but tested negative for SARS-CoV-2. This study aimed to shed light on this during the COVID-19 pandemic in Iran. METHODS In this cross-sectional study, a total of 1,002 patients with acute respiratory infection who had negative SARS-CoV-2 test results and referred to Valfajr Health Center, the National Collaborating Laboratory of Influenza and COVID-19 National Reference Laboratory at Pasteur Institute of Iran were recruited between January 2020 and January 2022. Nasopharyngeal and oropharyngeal swab samples were collected to detect 17 common respiratory viruses via TaqMan one-step real-time multiplex PCR. Demographic and clinical data of the participants were obtained from their electronic medical records. RESULTS In total, 218 samples (21.8%) were tested positive for at least one respiratory virus infection. Most of the common investigated respiratory viruses belonged to the years 2020 and 2022. The number of investigated patients in 2021 was few, which highlights the impact of health measures following the COVID-19 pandemic in Iran. Influenza A was the most common virus (5.8%), while adenovirus had the lowest prevalence (0.1%). Although the rate of respiratory virus infection was higher in men (24%) compared to women (19.3%), this difference was not statistically significant (P = 0.069). The prevalence of respiratory viruses had an inverse association with increasing age, with the highest rate (55.6%) observed in the age group below 2 years and the lowest rate (12.7%) in those above 65 years. CONCLUSION Our findings underscore the significance of adopting a comprehensive approach to respiratory infections detection and management. These results can be employed for the development of syndromic surveillance systems and implementation of the effective infection control measures. Furthermore, the results contribute to better understanding of the dynamics of respiratory viruses, both during pandemic periods and in non-pandemic contexts.
Collapse
Affiliation(s)
- Ali Maleki
- COVID-19 National Reference Laboratory, Pasteur Institute of Iran, Tehran, Iran
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Farah Bokharaei-Salim
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Mahsa Tavakoli
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | | | - Zahra Hosseini
- COVID-19 National Reference Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Setareh Kashanian
- COVID-19 National Reference Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Laya Farhan Asadi
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Salehi-Vaziri
- COVID-19 National Reference Laboratory, Pasteur Institute of Iran, Tehran, Iran.
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran.
| | - Fatemeh Fotouhi
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|