1
|
Mai KL, Pan WQ, Lin ZS, Wang Y, Yang ZF. Pathogenesis of influenza and SARS-CoV-2 co-infection at the extremes of age: decipher the ominous tales of immune vulnerability. ADVANCED BIOTECHNOLOGY 2025; 3:5. [PMID: 39883362 DOI: 10.1007/s44307-025-00057-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
The co-circulation of influenza and SARS-CoV-2 has led to co-infection events, primarily affecting children and older adults, who are at higher risk for severe disease. Although co-infection prevalence is relatively low, it is associated with worse outcomes compared to mono-infections. Previous studies have shown that the outcomes of co-infection depend on multiple factors, including viral interference, virus-host interaction and host response. Children and the elderly exhibit distinct patterns of antiviral response, which involve airway epithelium, mucociliary clearance, innate and adaptive immune cells, and inflammatory mediators. This review explores the pathogeneses of SARS-CoV-2 and influenza co-infection, focusing on the antiviral responses in children and the elderly. By comparing immature immunity in children and immune senescence in older adults, we aim to provide insights for the clinical management of severe co-infection cases.
Collapse
Affiliation(s)
- Kai-Lin Mai
- Henan University College of Medicine, Kaifeng, 475004, China
- School of Life Sciences, Henan University, Kaifeng, 475004, China
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei-Qi Pan
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zheng-Shi Lin
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Wang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Zi-Feng Yang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
2
|
Schlößer S, Ullrich AL, Modares NF, Schmitz MA, Schöneich J, Zhang K, Richter I, Robrahn L, Schraven S, Nagai JS, Haange SB, Jennings SAV, Clavel T, Rolle-Kampczyk U, Kiessling F, Costa IG, Muncan V, Repnik U, von Bergen M, Dupont A, Hornef MW. Salmonella infection accelerates postnatal maturation of the intestinal epithelium. Proc Natl Acad Sci U S A 2025; 122:e2403344122. [PMID: 39793046 PMCID: PMC11725846 DOI: 10.1073/pnas.2403344122] [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: 02/27/2024] [Accepted: 11/26/2024] [Indexed: 01/12/2025] Open
Abstract
Postnatal establishment of enteric metabolic, host-microbial and immune homeostasis is the result of precisely timed and tightly regulated developmental and adaptive processes. Here, we show that infection with the invasive enteropathogen Salmonella Typhimurium results in accelerated maturation of the neonatal epithelium with premature appearance of antimicrobial, metabolic, developmental, and regenerative features of the adult tissue. Using conditional Myd88-deficient mice, we identify the critical contribution of immune cell-derived mediators. Cytokine stimulation of neonatal intestinal epithelial stem cell organoids suggests a network of synergistic and antagonistic cytokine effects with a significant contribution of IL-22, IL-4/IL-13, TNF, and IL-6 to infection-induced enterocyte reprogramming. Our findings demonstrate that the infection-associated immune cell activation disrupts physiological postnatal tissue maturation and may thereby worsen clinical outcomes and alter the neonatal-adult transition.
Collapse
Grants
- SFB 1382 403224013 Deutsche Forschungsgemeinschaft (DFG)
- SFB 1382 403224013 Deutsche Forschungsgemeinschaft (DFG)
- SFB 1382 403224013 Deutsche Forschungsgemeinschaft (DFG)
- SFB/TRR359 491676693 Deutsche Forschungsgemeinschaft (DFG)
- SFB/TRR359 491676693 Deutsche Forschungsgemeinschaft (DFG)
- SPP2225 HO2236/18-1 Deutsche Forschungsgemeinschaft (DFG)
- SPP2389 DU 1803/2-1 Deutsche Forschungsgemeinschaft (DFG)
- DU-1803/1 Deutsche Forschungsgemeinschaft (DFG)
- eMed Consortia Fibromap Bundesministerium für Bildung und Forschung (BMBF)
- eMed Consortia Fibromap Bundesministerium für Bildung und Forschung (BMBF)
- START RWTH Aachen | Medizinische Fakultät, RWTH Aachen University (Faculty of Medicine, RWTH Aachen University)
- START RWTH Aachen | Medizinische Fakultät, RWTH Aachen University (Faculty of Medicine, RWTH Aachen University)
- Advanced Grant EarlyLife 101019157 EC | ERC | HORIZON EUROPE European Research Council (ERC)
- START RWTH Aachen | Medizinische Fakultät, RWTH Aachen University (Faculty of Medicine, RWTH Aachen University)
- START RWTH Aachen | Medizinische Fakultät, RWTH Aachen University (Faculty of Medicine, RWTH Aachen University)
- IRS seed fund RWTH Aachen University (RWTH Aachen)
- START RWTH Aachen | Medizinische Fakultät, RWTH Aachen University (Faculty of Medicine, RWTH Aachen University)
Collapse
Affiliation(s)
- Stefan Schlößer
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Anna-Lena Ullrich
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Nastaran Fazel Modares
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover30625, Germany
| | - Matthias A. Schmitz
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Johannes Schöneich
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Kaiyi Zhang
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Isabel Richter
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Laura Robrahn
- Institute for Computational Genomics, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
- Department of General, Visceral and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Sarah Schraven
- Institute for Experimental Molecular Imaging, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - James S. Nagai
- Institute for Computational Genomics, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Sven-Bastiaan Haange
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH—Helmholtz-Zentrum für Umweltforschung, Leipzig04318, Germany
| | - Susan A. V. Jennings
- Functional Microbiome Research Group, Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
- Euregional Microbiome Center, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH—Helmholtz-Zentrum für Umweltforschung, Leipzig04318, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Ivan G. Costa
- Institute for Computational Genomics, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC University of Amsterdam, Amsterdam1015 BK, The Netherlands
| | - Urska Repnik
- Central Microscopy, Christian Albrechts University, Kiel24118, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH—Helmholtz-Zentrum für Umweltforschung, Leipzig04318, Germany
| | - Aline Dupont
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover30625, Germany
| | - Mathias W. Hornef
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover30625, Germany
- Euregional Microbiome Center, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| |
Collapse
|
3
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Wu Z, Tien NTN, Bæk O, Zhong J, Klabunde B, Nguyen TT, Yen NTH, Long NP, Nguyen DN. Regulation of host metabolism and defense strategies to survive neonatal infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167482. [PMID: 39213794 DOI: 10.1016/j.bbadis.2024.167482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Two distinct defense strategies, disease resistance (DR) and disease tolerance (DT), enable a host to survive infectious diseases. Newborns, constrained by limited energy reserves, predominantly rely on DT to cope with infection. However, this approach may fail when pathogen levels surpass a critical threshold, prompting a shift to DR that can lead to dysregulated immune responses and sepsis. The mechanisms governing the interplay between DR and DT in newborns remain poorly understood. Here, we compare metabolic traits and defense strategies between survivors and non-survivors in Staphylococcus epidermidis (S. epidermidis)-infected preterm piglets, mimicking infection in preterm infants. Compared to non-survivors, survivors displayed elevated DR during the initial phase of infection, followed by stronger DT in later stages. In contrast, non-survivors showed clear signs of respiratory and metabolic acidosis and hyperglycemia, together with exaggerated inflammation and organ dysfunctions. Hepatic transcriptomics revealed a strong association between the DT phenotype and heightened oxidative phosphorylation in survivors, coupled with suppressed glycolysis and immune signaling. Plasma metabolomics confirmed the findings of metabolic regulations associated with DT phenotype in survivors. Our study suggests a significant association between the initial DR and subsequent DT, which collectively contributes to improved infection survival. The regulation of metabolic processes that optimize the timing and balance between DR and DT holds significant potential for developing novel therapeutic strategies for neonatal infection.
Collapse
Affiliation(s)
- Ziyuan Wu
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Ole Bæk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Jingren Zhong
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Björn Klabunde
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Tinh Thu Nguyen
- Department of Pediatrics, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
5
|
Martino D, Kresoje N, Amenyogbe N, Ben-Othman R, Cai B, Lo M, Idoko O, Odumade OA, Falsafi R, Blimkie TM, An A, Shannon CP, Montante S, Dhillon BK, Diray-Arce J, Ozonoff A, Smolen KK, Brinkman RR, McEnaney K, Angelidou A, Richmond P, Tebbutt SJ, Kampmann B, Levy O, Hancock REW, Lee AHY, Kollmann TR. DNA Methylation signatures underpinning blood neutrophil to lymphocyte ratio during first week of human life. Nat Commun 2024; 15:8167. [PMID: 39289350 PMCID: PMC11408723 DOI: 10.1038/s41467-024-52283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Understanding of newborn immune ontogeny in the first week of life will enable age-appropriate strategies for safeguarding vulnerable newborns against infectious diseases. Here we conducted an observational study exploring the immunological profile of infants longitudinally throughout their first week of life. Our Expanded Program on Immunization - Human Immunology Project Consortium (EPIC-HIPC) studies the epigenetic regulation of systemic immunity using small volumes of peripheral blood samples collected from West African neonates on days of life (DOL) 0, 1, 3, and 7. Genome-wide DNA methylation and single nucleotide polymorphism markers are examined alongside matched transcriptomic and flow cytometric data. Integrative analysis reveals that a core network of transcription factors mediates dynamic shifts in neutrophil-to-lymphocyte ratios (NLR), which are underpinned by cell-type specific methylation patterns in the two cell types. Genetic variants are associated with lower NLRs at birth, and healthy newborns with lower NLRs at birth are more likely to subsequently develop sepsis. These findings provide valuable insights into the early-life determinants of immune system development.
Collapse
Affiliation(s)
- David Martino
- The Kids Research Institute Australia, Perth, WA, Australia.
- University of Western Australia, Crawley, WA, Australia.
| | - Nina Kresoje
- The Kids Research Institute Australia, Perth, WA, Australia
| | - Nelly Amenyogbe
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Bing Cai
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Mandy Lo
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Olubukola Idoko
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Reza Falsafi
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Travis M Blimkie
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Andy An
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Casey P Shannon
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | | | - Bhavjinder K Dhillon
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Asimenia Angelidou
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neonatology, Beth Israel Deaconess Medical Centre, Boston, MA, USA
| | - Peter Richmond
- The Kids Research Institute Australia, Perth, WA, Australia
- University of Western Australia, Crawley, WA, Australia
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Beate Kampmann
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
- Centre for Global Health and Institute for International Health, Charite Universitatsmedizin, Berlin, Germany
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E W Hancock
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Tobias R Kollmann
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
6
|
Garvey M. Neonatal Infectious Disease: A Major Contributor to Infant Mortality Requiring Advances in Point-of-Care Diagnosis. Antibiotics (Basel) 2024; 13:877. [PMID: 39335050 PMCID: PMC11428345 DOI: 10.3390/antibiotics13090877] [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: 08/20/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Neonatal infectious disease continues to result in high rates of infant morbidity and mortality. Early- and late-onset disease represent difficult to detect and difficult to treat illnesses, particularly when antimicrobial resistant pathogens are present. Newborns are immunodeficient and are at increased risk of vertical and horizontal infection, with preterm infants increasingly susceptible. Additional risk factors associated with infection include prolonged use of a central catheter and/or ventilation, congenital abnormalities, admittance to intensive care units, and the use of broad-spectrum antibiotics. There is increasing recognition of the importance of the host microbiome and dysbiosis on neonatal infectious disease, including necrotising enterocolitis and sepsis in patients. Current diagnostic methods rely on blood culture, which is unreliable, time consuming, and can result in false negatives. There is a lack of accurate and reliable diagnostic tools available for the early detection of infectious disease in infants; therefore, efficient triage and treatment remains challenging. The application of biomarkers, machine learning, artificial intelligence, biosensors, and microfluidics technology, may offer improved diagnostic methodologies. Point-of-care devices, such diagnostic methodologies, may provide fast, reliable, and accurate diagnostic aids for neonatal patients. This review will discuss neonatal infectious disease as impacted by antimicrobial resistance and will highlight novel point-of-care diagnostic options.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
7
|
Saager ES, van Stigt AH, Lerkvaleekul B, Lutter L, Hellinga AH, van der Wal MM, Bont LJ, Leusen JH, van’t Land B, van Wijk F. Human breastmilk memory T cells throughout lactation manifest activated tissue-oriented profile with prominent regulation. JCI Insight 2024; 9:e181788. [PMID: 39435660 PMCID: PMC11530127 DOI: 10.1172/jci.insight.181788] [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: 10/23/2024] Open
Abstract
Breastfeeding provides important immunological benefits to the neonate, but how the different immunoactive components in breastmilk contribute to immunity remains poorly understood. Here, we characterized human breastmilk T cells using single-cell RNA-Seq and flow cytometry. Breastmilk contained predominantly memory T cells, with expression of immune signaling genes, high proliferation, and an effector Th1/cytotoxic profile with high cytokine production capacities. Elevated activation was balanced by an enriched Treg population and immune regulatory markers in conventional memory T cells. Gene and surface expression of tissue-residency markers indicate that breastmilk T cells represented tissue-adapted rather than circulatory T cells. In addition, breastmilk T cells had a broad homing profile and higher activation markers in these migratory subsets. The partly overlapping transcriptome profile between breastmilk and breast tissue T cells, particularly cytotoxic T cells, might support a role in local immune defense in the mammary gland. However, unique features of breastmilk, such as Tregs, might imply an additional role in neonatal immune support. We found some correlations between the breastmilk T cell profile and clinical parameters, most notably with maternal and household factors. Together, our data suggest that breastmilk contains an adapted T cell population that exerts their function in specific tissue sites.
Collapse
Affiliation(s)
- Elise S. Saager
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Arthur H. van Stigt
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Butstabong Lerkvaleekul
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lisanne Lutter
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- Department of Pathology, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Anneke H. Hellinga
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - M. Marlot van der Wal
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Louis J. Bont
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital/University Medical Center Utrecht, Utrecht, Netherlands
- ReSViNET foundation, Zeist, Netherlands
| | - Jeanette H.W. Leusen
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Belinda van’t Land
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- CoE Immunology, Danone Global Research & Innovation Center, Utrecht, Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | | |
Collapse
|
8
|
Adiga V, Bindhu H, Ahmed A, Chetan Kumar N, Tripathi H, D’Souza G, Dias M, Shivalingaiah S, Rao S, K N S, Hawrylowicz C, Dwarkanath P, Vyakarnam A. Immune profiling reveals umbilical cord blood mononuclear cells from South India display an IL-8 dominant, CXCL-10 deficient polyfunctional monocyte response to pathogen-associated molecular patterns that is distinct from adult blood cells. Clin Exp Immunol 2024; 217:263-278. [PMID: 38695079 PMCID: PMC11310697 DOI: 10.1093/cei/uxae034] [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: 11/30/2023] [Revised: 02/28/2024] [Accepted: 05/14/2024] [Indexed: 08/10/2024] Open
Abstract
Neonate responses to pathogen-associated molecular patterns (PAMPS) differ from adults; such understanding is poor in Indian neonates, despite recognized significant infectious risk. Immune profiling analysis was undertaken of 10 secreted mediators contextualized with cellular source induced by six PAMPs in umbilical cord (CB; n = 21) and adult-blood (PBMC; n = 14) from a tertiary care hospital in South India. Differential cytokine expression analysis (minimum log2-fold difference; adj P-value < 0.05) identified bacterial PAMPs induced higher concentrations of IL-1β, IL-10, TNF-α in adults versus IL-8, GM-CSF, IFN-γ, and IL-2 in CB. CB responded to poly I:C and SARS-CoV-2 lysate with a dominant IL-8 response, whereas in PBMC, CXCL-10 dominated poly I:C, but not SARS-CoV-2, responses, highlighting potential IL-8 importance, in the absence of Type I Interferons, in antiviral CB immunity. Candida albicans was the only PAMP to uniformly induce higher secretion of effectors in CB. The predominant source of IL-8/IL-6/TNF-α/IL-1β in both CB and PBMC was polyfunctional monocytes and IFN-γ/IL-2/IL-17 from innate lymphocytes. Correlation matrix analyses revealed IL-8 to be the most differentially regulated, correlating positively in CB versus negatively in PBMC with IL-6, GM-CSF, IFN-γ, IL-2, consistent with more negatively regulated cytokine modules in adults, potentially linked to higher anti-inflammatory IL-10. Cord and adult blood from India respond robustly to PAMPs with unique effector combinations. These data provide a strong foundation to monitor, explore, mechanisms that regulate such immunity during the life course, an area of significant global health importance given infection-related infant mortality incidence.
Collapse
Affiliation(s)
- Vasista Adiga
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Hima Bindhu
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Asma Ahmed
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Nirutha Chetan Kumar
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Himanshu Tripathi
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - George D’Souza
- Department of Pulmonary Medicine, St. John’s Medical College, Bangalore, India
| | - Mary Dias
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | | | - Srishti Rao
- Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Shanti K N
- Department of Biotechnology, PES University, Bangalore, Karnataka, India
| | - Catherine Hawrylowicz
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| | - Pratibha Dwarkanath
- Division of Nutrition, St. John’s Research Institute, Bangalore, Karnataka, India
| | - Annapurna Vyakarnam
- Human Immunology Laboratory, Division of Infectious Diseases, St. John’s Research Institute, Bangalore, Karnataka, India
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Science & Medicine, King’s College, London, UK
| |
Collapse
|
9
|
Strunk T, Molloy EJ, Mishra A, Bhutta ZA. Neonatal bacterial sepsis. Lancet 2024; 404:277-293. [PMID: 38944044 DOI: 10.1016/s0140-6736(24)00495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/06/2024] [Accepted: 03/07/2024] [Indexed: 07/01/2024]
Abstract
Neonatal sepsis remains one of the key challenges of neonatal medicine, and together with preterm birth, causes almost 50% of all deaths globally for children younger than 5 years. Compared with advances achieved for other serious neonatal and early childhood conditions globally, progress in reducing neonatal sepsis has been much slower, especially in low-resource settings that have the highest burden of neonatal sepsis morbidity and mortality. By contrast to sepsis in older patients, there is no universally accepted neonatal sepsis definition. This poses substantial challenges in clinical practice, research, and health-care management, and has direct practical implications, such as diagnostic inconsistency, heterogeneous data collection and surveillance, and inappropriate treatment, health-resource allocation, and education. As the clinical manifestation of neonatal sepsis is frequently non-specific and the current diagnostic standard blood culture has performance limitations, new improved diagnostic techniques are required to guide appropriate and warranted antimicrobial treatment. Although antimicrobial therapy and supportive care continue as principal components of neonatal sepsis therapy, refining basic neonatal care to prevent sepsis through education and quality improvement initiatives remains paramount.
Collapse
Affiliation(s)
- Tobias Strunk
- Neonatal Directorate, King Edward Memorial Hospital, Child and Adolescent Health Service, Perth, WA, Australia; Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College, University of Dublin and Trinity Research in Childhood Centre, Dublin, Ireland; Children's Health Hospital at Tallaght, Tallaght University Hospital, Dublin, Ireland; Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland; Neonatology, Children's Health Hospital at Crumlin, Dublin, Ireland; Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Archita Mishra
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Zulfiqar A Bhutta
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada; Institute for Global Health and Development, The Aga Khan University South-Central Asia, Karachi, Pakistan
| |
Collapse
|
10
|
Joana Alves M, Browe BM, Carolina Rodrigues Dias A, Torres JM, Zaza G, Bangudi S, Blackburn J, Wang W, de Araujo Fernandes-Junior S, Fadda P, Toland A, Baer LA, Stanford KI, Czeisler C, Garcia AJ, Javier Otero J. Metabolic trade-offs in Neonatal sepsis triggered by TLR4 and TLR1/2 ligands result in unique dysfunctions in neural breathing circuits. Brain Behav Immun 2024; 119:333-350. [PMID: 38561095 DOI: 10.1016/j.bbi.2024.03.027] [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: 11/14/2023] [Revised: 03/05/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Neonatal sepsis remains one of the leading causes of mortality in newborns. Several brainstem-regulated physiological processes undergo disruption during neonatal sepsis. Mechanistic knowledge gaps exist at the interplay between metabolism and immune activation to brainstem neural circuits and pertinent physiological functions in neonates. To delineate this association, we induced systemic inflammation either by TLR4 (LPS) or TLR1/2 (PAM3CSK4) ligand administration in postnatal day 5 mice (PD5). Our findings show that LPS and PAM3CSK4 evoke substantial changes in respiration and metabolism. Physiological trade-offs led to hypometabolic-hypothermic responses due to LPS, but not PAM3CSK4, whereas to both TLR ligands blunted respiratory chemoreflexes. Neuroinflammatory pathways modulation in brainstem showed more robust effects in LPS than PAM3CSK4. Brainstem neurons, microglia, and astrocyte gene expression analyses showed unique responses to TLR ligands. PAM3CSK4 did not significantly modulate gene expression changes in GLAST-1 positive brainstem astrocytes. PD5 pups receiving PAM3CSK4 failed to maintain a prolonged metabolic state repression, which correlated to enhanced gasping latency and impaired autoresuscitation during anoxic chemoreflex challenges. In contrast, LPS administered pups showed no significant changes in anoxic chemoreflex. Electrophysiological studies from brainstem slices prepared from pups exposed to either TLR4 or PAM3CSK4 showed compromised transmission between preBötzinger complex and Hypoglossal as an exclusive response to the TLR1/2 ligand. Spatial gene expression analysis demonstrated a region-specific modulation of PAM3CSK4 within the raphe nucleus relative to other anatomical sites evaluated. Our findings suggest that metabolic changes due to inflammation might be a crucial tolerance mechanism for neonatal sepsis preserving neural control of breathing.
Collapse
Affiliation(s)
- Michele Joana Alves
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Brigitte M Browe
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Neuroscience Institute, The University of Chicago, Chicago, IL, United States
| | - Ana Carolina Rodrigues Dias
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Juliet M Torres
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Giuliana Zaza
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Suzy Bangudi
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Jessica Blackburn
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Wesley Wang
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | | | - Paolo Fadda
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Amanda Toland
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States; Department of Cancer Biology and Genetics and Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Lisa A Baer
- Department of Cancer Biology and Genetics and Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Catherine Czeisler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Alfredo J Garcia
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Neuroscience Institute, The University of Chicago, Chicago, IL, United States.
| | - José Javier Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, United States.
| |
Collapse
|
11
|
Borghesi A. Life-threatening infections in human newborns: Reconciling age-specific vulnerability and interindividual variability. Cell Immunol 2024; 397-398:104807. [PMID: 38232634 DOI: 10.1016/j.cellimm.2024.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
In humans, the interindividual variability of clinical outcome following exposure to a microorganism is immense, ranging from silent infection to life-threatening disease. Age-specific immune responses partially account for the high incidence of infection during the first 28 days of life and the related high mortality at population level. However, the occurrence of life-threatening disease in individual newborns remains unexplained. By contrast, inborn errors of immunity and their immune phenocopies are increasingly being discovered in children and adults with life-threatening viral, bacterial, mycobacterial and fungal infections. There is a need for convergence between the fields of neonatal immunology, with its in-depth population-wide characterization of newborn-specific immune responses, and clinical immunology, with its investigations of infections in patients at the cellular and molecular levels, to facilitate identification of the mechanisms of susceptibility to infection in individual newborns and the design of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, EU, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| |
Collapse
|
12
|
Logsdon AF, Erickson MA, Herbert MJ, Noonan C, Foresi BD, Qiu J, Lim YP, Banks WA, Stonestreet BS. Inter-alpha inhibitor proteins attenuate lipopolysaccharide-induced blood-brain barrier disruption in neonatal mice. Exp Neurol 2023; 370:114563. [PMID: 37806514 DOI: 10.1016/j.expneurol.2023.114563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/21/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
There is a paucity of information regarding efficacious pharmacological neuroprotective strategies to attenuate or reduce brain injury in neonates. Lipopolysaccharide (LPS) disrupts blood-brain barrier (BBB) function in adult rodents and increases inflammation in adults and neonates. Human blood-derived Inter-alpha Inhibitor Proteins (IAIPs) are neuroprotective, improve neonatal survival after LPS, and attenuate LPS-induced disruption of the BBB in adult male mice. We hypothesized that LPS also disrupts the function of the BBB in neonatal mice and that IAIPs attenuate the LPS-induced BBB disruption in male and female neonatal mice. IAIPs were administered to neonatal mice after LPS and BBB permeability quantified with intravenous 14C-sucrose and 99mTc-albumin. Although repeated high doses (3 mg/kg) of LPS in neonates resulted in high mortality rates and a robust increase in BBB permeability, repeated lower doses (1 mg/kg) of LPS resulted in lower mortality rates and disruption of the BBB in both male and female neonates. IAIP treatment attenuated disruption of the BBB similarly to sucrose and albumin after exposure to low-dose LPS in neonatal mice. Exposure to low-dose LPS elevated IAIP concentrations in blood, but it did not appear to increase the systemic levels of Pre-alpha inhibitor (PaI), one of the family members of the IAIPs that contains heavy chain 3. We conclude that IAIPs attenuate LPS-related disruption of the BBB in both male and female neonatal mice.
Collapse
Affiliation(s)
- Aric F Logsdon
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Melanie J Herbert
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Cassidy Noonan
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Brian D Foresi
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI 02903, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02903, USA; Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Barbara S Stonestreet
- The Alpert Medical School of Brown University, Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI 02905, USA
| |
Collapse
|
13
|
Sedney CJ, Harvill ET. Making friends: active selection of symbionts and rejection of pathogens by the neonatal immune system. Front Immunol 2023; 14:1287518. [PMID: 38054002 PMCID: PMC10694279 DOI: 10.3389/fimmu.2023.1287518] [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: 09/01/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The neonatal immune system is generally viewed as deficient compared to adults, often attributed to its incomplete development. This view is reinforced by the extraordinary sensitivity and susceptibility of neonates to certain pathogens. Examination of the basis for this susceptibility has characterized neonatal immunity as skewed strongly toward anti-inflammatory responses, which are interpreted as the lack of full development of the strong inflammatory responses observed in adults. Here we examine the alternative explanation that neonatal immune responses are generally complete in healthy newborns but evolved and adapted to very different functions than adult immunity. Adult immunity is primarily aimed at controlling pathogens that invade the holobiont, with substantial competition and protection conferred by resident microbiota. Rather than simply repelling new invaders, the immediate and critical challenge of the neonatal immune system during the sudden transition from near sterility to microbe-rich world is the assimilation of a complex microbiota to generate a stable and healthy holobiont. This alternative view of the role of the neonatal immune system both explains its strong anti-inflammatory bias and provides a different perspective on its other unique aspects. Here we discuss recent work exploring the initial contact of newborns with microbes and their interactions with neonatal immune responses, contrasting these alternative perspectives. Understanding how the need to rapidly acquire a highly complex and rich microbiota of commensals affects interactions between the neonatal immune system and both commensals and pathogens will allow more targeted and effective collaboration with this system to quickly achieve a more disease-resistant holobiont.
Collapse
Affiliation(s)
- Colleen J. Sedney
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | | |
Collapse
|
14
|
de Jong R, Tenbrock K, Ohl K. New Insights in Immunometabolism in Neonatal Monocytes and Macrophages in Health and Disease. Int J Mol Sci 2023; 24:14173. [PMID: 37762476 PMCID: PMC10531550 DOI: 10.3390/ijms241814173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
It is well established that the neonatal immune system is different from the adult immune system. A major task of the neonatal immune system is to bridge the achievement of tolerance towards harmless antigens and commensal bacteria while providing protection against pathogens. This is highly important because neonates are immunologically challenged directly after birth by a rigorous change from a semi-allogeneic sterile environment into a world rich with microbes. A so called disease tolerogenic state is typical for neonates and is anticipated to prevent immunopathological damage potentially at the cost of uncontrolled pathogen proliferation. As a consequence, neonates are more susceptible than adults to life-threatening infections. At the basis of a well-functioning immune response, both for adults and neonates, innate immune cells such as monocytes and monocyte-derived macrophages play an essential role. A well-responsive monocyte will alter its cellular metabolism to subsequently induce certain immune effector function, a process which is called immunometabolism. Immunometabolism has received extensive attention in the last decade; however, it has not been broadly studied in neonates. This review focuses on carbohydrate metabolism in monocytes and macrophages in neonates. We will exhibit pathways involving glycolysis, the tricarboxylic acid (TCA) cycle and oxidative phosphorylation and their role in shaping neonates' immune systems to a favorable tolerogenic state. More insight into these pathways will elucidate potential treatments targets in life-threatening conditions including neonatal sepsis or expose potential targets which can be used to induce tolerance in conditions where tolerance is harmfully impaired such as in autoimmune diseases.
Collapse
Affiliation(s)
| | - Klaus Tenbrock
- Department of Pediatrics, RWTH Aachen University, 52074 Aachen, Germany; (R.d.J.); (K.O.)
| | | |
Collapse
|
15
|
Andualem H, Hollams E, Kollmann TR, Amenyogbe N. BCG-Induced Immune Training: Interplay between Trained Immunity and Emergency Granulopoiesis. J Mol Biol 2023; 435:168169. [PMID: 37263392 PMCID: PMC11688642 DOI: 10.1016/j.jmb.2023.168169] [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: 02/28/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
Bacille Calmette-Guérin (BCG) is the most commonly administered vaccine in human history. The medical application of BCG extends far beyond the fight against tuberculosis. Despite its stellar medical record over 100 years, insight into how BCG provides this vast range of benefits is largely limited, both for its pathogen-specific (tuberculosis) as well as pathogen-agnostic (other infections, autoimmunity, allergies, and cancer) effects. Trained immunity and emergency granulopoiesis have been identified as mediating BCG's pathogen-agnostic effects, for which some of the molecular mechanisms have been delineated. Upon review of the existing evidence, we postulate that emergency granulopoiesis and trained immunity are a continuum of the same effect cascade. In this context, we highlight that BCG's pathogen-agnostic benefits could be optimized by taking advantage of the age of the recipient and route of BCG administration.
Collapse
Affiliation(s)
- Henok Andualem
- Department of Medical Laboratory Science, College of Health Science, Debre Tabor University, Ethiopia.
| | - Elysia Hollams
- Telethon Kids Institute, Perth, Western Australia, Australia
| | | | - Nelly Amenyogbe
- Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
16
|
Pietrasanta C, Conti MG. Editorial: Neonatal infections and the developing neonatal immune system: current evidence and research gaps to fill. Front Pediatr 2023; 11:1243752. [PMID: 37528874 PMCID: PMC10389776 DOI: 10.3389/fped.2023.1243752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Affiliation(s)
- Carlo Pietrasanta
- NICU, Department of Woman, Child and Newborn, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
17
|
Giles ML, Way SS, Marchant A, Aghaepour N, James T, Schaltz-Buchholzer F, Zazara D, Arck P, Kollmann TR. Maternal Vaccination to Prevent Adverse Pregnancy Outcomes: An Underutilized Molecular Immunological Intervention? J Mol Biol 2023; 435:168097. [PMID: 37080422 PMCID: PMC11533213 DOI: 10.1016/j.jmb.2023.168097] [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: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
Adverse pregnancy outcomes including maternal mortality, stillbirth, preterm birth, intrauterine growth restriction cause millions of deaths each year. More effective interventions are urgently needed. Maternal immunization could be one such intervention protecting the mother and newborn from infection through its pathogen-specific effects. However, many adverse pregnancy outcomes are not directly linked to the infectious pathogens targeted by existing maternal vaccines but rather are linked to pathological inflammation unfolding during pregnancy. The underlying pathogenesis driving such unfavourable outcomes have only partially been elucidated but appear to relate to altered immune regulation by innate as well as adaptive immune responses, ultimately leading to aberrant maternal immune activation. Maternal immunization, like all immunization, impacts the immune system beyond pathogen-specific immunity. This raises the possibility that maternal vaccination could potentially be utilised as a pathogen-agnostic immune modulatory intervention to redirect abnormal immune trajectories towards a more favourable phenotype providing pregnancy protection. In this review we describe the epidemiological evidence surrounding this hypothesis, along with the mechanistic plausibility and present a possible path forward to accelerate addressing the urgent need of adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | - Sing Sing Way
- Center for Inflammation and Tolerance; Cincinnati Children's Hospital, Cincinnati, USA
| | | | - Nima Aghaepour
- Stanford University School of Medicine, Stanford, CA, USA
| | - Tomin James
- Stanford University School of Medicine, Stanford, CA, USA
| | | | - Dimitra Zazara
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Petra Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | | |
Collapse
|
18
|
Sedney CJ, Harvill ET. The Neonatal Immune System and Respiratory Pathogens. Microorganisms 2023; 11:1597. [PMID: 37375099 PMCID: PMC10301501 DOI: 10.3390/microorganisms11061597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Neonates are more susceptible to some pathogens, particularly those that cause infection in the respiratory tract. This is often attributed to an incompletely developed immune system, but recent work demonstrates effective neonatal immune responses to some infection. The emerging view is that neonates have a distinctly different immune response that is well-adapted to deal with unique immunological challenges of the transition from a relatively sterile uterus to a microbe-rich world, tending to suppress potentially dangerous inflammatory responses. Problematically, few animal models allow a mechanistic examination of the roles and effects of various immune functions in this critical transition period. This limits our understanding of neonatal immunity, and therefore our ability to rationally design and develop vaccines and therapeutics to best protect newborns. This review summarizes what is known of the neonatal immune system, focusing on protection against respiratory pathogens and describes challenges of various animal models. Highlighting recent advances in the mouse model, we identify knowledge gaps to be addressed.
Collapse
Affiliation(s)
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
19
|
Cho J, Han SC, Ho Hwang J, Song J. Characterization of immune development of fetal and early-life of minipigs. Int Immunopharmacol 2023; 120:110310. [PMID: 37196561 DOI: 10.1016/j.intimp.2023.110310] [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: 03/02/2023] [Revised: 04/27/2023] [Accepted: 05/06/2023] [Indexed: 05/19/2023]
Abstract
Fetal and child's immune systems differ from those of adults. Developing immune systems exhibit increased or decreased sensitivity to drugs, infection, or toxicants compared to adult immune systems. Understanding fetal and neonatal immune systems will help predict toxicity or the pathogenesis or prognosis of diseases. In this study, we evaluated whether the innate and adaptive immune system of fetal and young minipigs could respond to external stimuli compared to a medium-treated group and analyzed several immunological parameters for developmental immunotoxicity according to developmental stages. We performed a hematological analysis of fetal cord bloods and the bloods of neonatal and 4-week-old piglets. Splenocytes were isolated at each developmental stage and treated with lipopolysaccharide (LPS), R848, and concanavalin A (ConA). Various cytokines were measured in the cell supernatants. Total antibody production was also evaluated in serum. The percentage of lymphocytes was dominant in gestational weeks (GW) 10 and 12 and started to decrease from postnatal day (PND) 0. From PND0, the percentage of neutrophils increased. Interleukin (IL)-1β, IL-6, and interferon (IFN)-α were induced from GW10 in response to LPS and R848 stimulation. Th1 cytokine induction was detected from PND0 upon ConA stimulation, whereas Th2 cytokine release was observed from GW10. IgM and IgG production was sustained at low levels at fetal stages and was significantly increased after birth. This study reconfirmed that the fetal immune system could respond to external stimuli and that hematological analysis, cytokine evaluation, and antibody subclass measurement can be useful parameters for developmental immunotoxicity using minipigs.
Collapse
Affiliation(s)
- Jeonghee Cho
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea; Graduate School of Konyang University of Bioconvergence, Department of Bio-Non-Clinical Science, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Republic of Korea
| | - Su-Cheol Han
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea.
| | - Jeongah Song
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea.
| |
Collapse
|
20
|
Torow N, Hand TW, Hornef MW. Programmed and environmental determinants driving neonatal mucosal immune development. Immunity 2023; 56:485-499. [PMID: 36921575 PMCID: PMC10079302 DOI: 10.1016/j.immuni.2023.02.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023]
Abstract
The mucosal immune system of neonates goes through successive, non-redundant phases that support the developmental needs of the infant and ultimately establish immune homeostasis. These phases are informed by environmental cues, including dietary and microbial stimuli, but also evolutionary developmental programming that functions independently of external stimuli. The immune response to exogenous stimuli is tightly regulated during early life; thresholds are set within this neonatal "window of opportunity" that govern how the immune system will respond to diet, the microbiota, and pathogenic microorganisms in the future. Thus, changes in early-life exposure, such as breastfeeding or environmental and microbial stimuli, influence immunological and metabolic homeostasis and the risk of developing diseases such as asthma/allergy and obesity.
Collapse
Affiliation(s)
- Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Timothy W Hand
- Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA.
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| |
Collapse
|
21
|
França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics (Basel) 2023; 12:antibiotics12030554. [PMID: 36978421 PMCID: PMC10044083 DOI: 10.3390/antibiotics12030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.
Collapse
Affiliation(s)
- Angela França
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
| |
Collapse
|
22
|
Sedney CJ, Caulfield A, Dewan KK, Blas-Machado U, Callender M, Manley NR, Harvill ET. Novel murine model reveals an early role for pertussis toxin in disrupting neonatal immunity to Bordetella pertussis. Front Immunol 2023; 14:1125794. [PMID: 36855631 PMCID: PMC9968397 DOI: 10.3389/fimmu.2023.1125794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
The increased susceptibility of neonates to specific pathogens has previously been attributed to an underdeveloped immune system. More recent data suggest neonates have effective protection against most pathogens but are particularly susceptible to those that target immune functions specific to neonates. Bordetella pertussis (Bp), the causative agent of "whooping cough", causes more serious disease in infants attributed to its production of pertussis toxin (PTx), although the neonate-specific immune functions it targets remain unknown. Problematically, the rapid development of adult immunity in mice has confounded our ability to study interactions of the neonatal immune system and its components, such as virtual memory T cells which are prominent prior to the maturation of the thymus. Here, we examine the rapid change in susceptibility of young mice and define a period from five- to eight-days-old during which mice are much more susceptible to Bp than mice even a couple days older. These more narrowly defined "neonatal" mice display significantly increased susceptibility to wild type Bp but very rapidly and effectively respond to and control Bp lacking PTx, more rapidly even than adult mice. Thus, PTx efficiently blocks some very effective form(s) of neonatal protective immunity, potentially providing a tool to better understand the neonatal immune system. The rapid clearance of the PTx mutant correlates with the early accumulation of neutrophils and T cells and suggests a role for PTx in disrupting their accumulation. These results demonstrate a striking age-dependent response to Bp, define an early age of extreme susceptibility to Bp, and demonstrate that the neonatal response can be more efficient than the adult response in eliminating bacteria from the lungs, but these neonatal functions are substantially blocked by PTx. This refined definition of "neonatal" mice may be useful in the study of other pathogens that primarily infect neonates, and PTx may prove a particularly valuable tool for probing the poorly understood neonatal immune system.
Collapse
Affiliation(s)
- Colleen J. Sedney
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Amanda Caulfield
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Kaylan K. Dewan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Uriel Blas-Machado
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Maiya Callender
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Nancy R. Manley
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
23
|
Manches O, Um K, Boudier A, Maddouri Y, Lyon-Caen S, Bayat S, Slama R, Philippat C, Siroux V, Chaperot L. Maternal imprinting and determinants of neonates' immune function in the SEPAGES mother-child cohort. Front Immunol 2023; 14:1136749. [PMID: 37081891 PMCID: PMC10111372 DOI: 10.3389/fimmu.2023.1136749] [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: 01/03/2023] [Accepted: 03/13/2023] [Indexed: 04/22/2023] Open
Abstract
Introduction Immune function in pregnancy is influenced by host-specific and environmental factors. This may impact fetal immune development, but the link between maternal and neonatal immune function is still poorly characterized. Here, we investigate the relationship between maternal and neonatal immune function, and identify factors affecting the association between maternal and child cytokine secretion. Methods In the French prospective cohort SEPAGES, blood samples were obtained from pregnant women (n=322) at gestational week 20 ± 4 and from their child at birth (n=156). Maternal and cord blood cytokine and chemokine (CK) levels were measured at baseline in all subjects and after T cell or dendritic cell activation with phytohemagglutinin or R848 (in total 29 and 27 measures in maternal and cord blood samples, respectively). Associations between environmental, individual factors and CK level were estimated by linear regression modeling. The maternal-cord blood CK relations were assessed by Pearson correlation and regression models. Results We observed that pregnant women and neonates displayed specific CK secretion profiles in the innate and adaptive compartments at baseline and upon activation. Activation of T cells in cord blood induced high levels of IL-2, but low levels of IFNγ, IL-13 or IL-10, in comparison to maternal blood samples. Elsewhere, neonatal innate immune responses were characterized by low production of IFNα, while productions of IL-1β, IL-6, IL-8, IL-10 and TNFα were higher than maternal responses. Strong correlations were observed between most CK after activation in maternal and cord blood samples. Strikingly, a statistical association between global mother and child cytokine profiles was evidenced. Correlations were observed between some individual CK of pregnant women and their children, both at baseline (MCP1, RANTES) and after activation with R848 (IL-6, IL-8 and IL-10). We looked for factors which could influence cytokine secretion in maternal or cord blood, and found that leucocyte counts, maternal age, pre-conception BMI, smoking and season were associated with the levels of several CK in mothers or children. Discussion Our study reveals in utero immune imprinting influencing immune responses in infants, opening the way to investigate the mechanisms responsible for this imprinting. Whether such influences have long lasting effects on children health warrants further investigation.
Collapse
Affiliation(s)
- Olivier Manches
- EFS, Recherche et Développement, Grenoble, France
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Khémary Um
- EFS, Recherche et Développement, Grenoble, France
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Anne Boudier
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
- Department of Pulmonology and Physiology, CHU Grenoble-Alpes, Grenoble, France
| | - Yasmina Maddouri
- EFS, Recherche et Développement, Grenoble, France
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Sarah Lyon-Caen
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Sam Bayat
- Department of Pulmonology and Physiology, CHU Grenoble-Alpes, Grenoble, France
| | - Rémy Slama
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Claire Philippat
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Valérie Siroux
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Chaperot
- EFS, Recherche et Développement, Grenoble, France
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR, Institute for Advanced Biosciences, Grenoble, France
- *Correspondence: Laurence Chaperot,
| |
Collapse
|
24
|
Kowalczyk P, Kaczyńska K, Kleczkowska P, Bukowska-Ośko I, Kramkowski K, Sulejczak D. The Lactoferrin Phenomenon-A Miracle Molecule. Molecules 2022; 27:2941. [PMID: 35566292 PMCID: PMC9104648 DOI: 10.3390/molecules27092941] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 01/11/2023] Open
Abstract
Numerous harmful factors that affect the human body from birth to old age cause many disturbances, e.g., in the structure of the genome, inducing cell apoptosis and their degeneration, which leads to the development of many diseases, including cancer. Among the factors leading to pathological processes, microbes, viruses, gene dysregulation and immune system disorders have been described. The function of a protective agent may be played by lactoferrin as a "miracle molecule", an endogenous protein with a number of favorable antimicrobial, antiviral, antioxidant, immunostimulatory and binding DNA properties. The purpose of this article is to present the broad spectrum of properties and the role that lactoferrin plays in protecting human cells at all stages of life.
Collapse
Affiliation(s)
- Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland;
| | - Patrycja Kleczkowska
- Maria Sklodowska-Curie, Medical Academy in Warsaw, Solidarności 12 St., 03-411 Warsaw, Poland
- Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163 Warsaw, Poland
| | - Iwona Bukowska-Ośko
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, Pawinskiego 3c St., 02-106 Warsaw, Poland;
| | - Karol Kramkowski
- Department of Physical Chemistry, Medical University of Bialystok, Kilińskiego 1 St., 15-089 Bialystok, Poland;
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
25
|
Hibbert J, Strunk T, Nathan E, Prosser A, Doherty D, Simmer K, Richmond P, Burgner D, Currie A. Composition of early life leukocyte populations in preterm infants with and without late-onset sepsis. PLoS One 2022; 17:e0264768. [PMID: 35235604 PMCID: PMC8890632 DOI: 10.1371/journal.pone.0264768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Composition of leukocyte populations in the first month of life remains incompletely characterised, particularly in preterm infants who go on to develop late-onset sepsis (LOS). AIM To characterise and compare leukocyte populations in preterm infants with and without LOS during the first month of life. STUDY DESIGN Single-centre prospective observational cohort study. PARTICIPANTS Infants born <30 weeks gestational age (GA). OUTCOME MEASURES Peripheral blood samples were collected at 1, 7, 14, 21 and 28 days of life. Leukocyte populations were characterised using 5-fluorophore-6-marker flow cytometry. Absolute leukocyte counts and frequency of total CD45+ leukocytes of each population were adjusted for GA, birth weight z-scores, sex and total leukocyte count. RESULTS Of 119 preterm infants enrolled, 43 (36%) had confirmed or clinical LOS, with a median onset at 13 days (range 6-26). Compared to infants without LOS, the adjusted counts and frequency of neutrophils, basophils and non-cytotoxic T lymphocytes were generally lower and immature granulocytes were higher over the first month of life in infants who developed LOS. Specific time point comparisons identified lower adjusted neutrophil counts on the first day of life in those infants who developed LOS more than a week later, compared to those without LOS, albeit levels were within the normal age-adjusted range. Non-cytotoxic T lymphocyte counts and/or frequencies were lower in infants following LOS on days 21 and 28 when compared to those who did not develop LOS. CONCLUSION Changes in non-cytotoxic T lymphocytes occurred following LOS suggesting sepsis-induced immune suppression.
Collapse
Affiliation(s)
- Julie Hibbert
- Neonatal Directorate, Child and Adolescent Health Service, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Tobias Strunk
- Neonatal Directorate, Child and Adolescent Health Service, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Elizabeth Nathan
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Amy Prosser
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Dorota Doherty
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Karen Simmer
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Peter Richmond
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - David Burgner
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Currie
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
- Centre for Molecular Medicine and Innovation Therapeutics, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
26
|
Look Who's Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis. Int J Mol Sci 2022; 23:ijms23020860. [PMID: 35055041 PMCID: PMC8775791 DOI: 10.3390/ijms23020860] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.
Collapse
|
27
|
Scanlon KM, Chen L, Carbonetti NH. Pertussis Toxin Promotes Pulmonary Hypertension in an Infant Mouse Model of Bordetella pertussis Infection. J Infect Dis 2022; 225:172-176. [PMID: 34145457 PMCID: PMC8730479 DOI: 10.1093/infdis/jiab325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
Pertussis, caused by Bordetella pertussis, is a reemerging disease that can produce severe disease manifestations in infants, including pulmonary hypertension (PH). B. pertussis-induced PH is a major risk factor for infection-induced death, but the molecular mechanisms promoting PH are unknown and there is no effective treatment. We examined B. pertussis-induced PH in infant and adult mouse models of pertussis by Fulton index, right heart catheterization, or Doppler echocardiogram. Our results demonstrate that B. pertussis-induced PH is age related and dependent on the expression of pertussis toxin by the bacterium. Hence, pertussis toxin-targeting treatments may ameliorate PH and fatal infant infection.
Collapse
Affiliation(s)
- Karen M Scanlon
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ling Chen
- Department of Physiology and Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nicholas H Carbonetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Jašarević E, Hill EM, Kane PJ, Rutt L, Gyles T, Folts L, Rock KD, Howard CD, Morrison KE, Ravel J, Bale TL. The composition of human vaginal microbiota transferred at birth affects offspring health in a mouse model. Nat Commun 2021; 12:6289. [PMID: 34725359 PMCID: PMC8560944 DOI: 10.1038/s41467-021-26634-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Newborns are colonized by maternal microbiota that is essential for offspring health and development. The composition of these pioneer communities exhibits individual differences, but the importance of this early-life heterogeneity to health outcomes is not understood. Here we validate a human microbiota-associated model in which fetal mice are cesarean delivered and gavaged with defined human vaginal microbial communities. This model replicates the inoculation that occurs during vaginal birth and reveals lasting effects on offspring metabolism, immunity, and the brain in a community-specific manner. This microbial effect is amplified by prior gestation in a maternal obesogenic or vaginal dysbiotic environment where placental and fetal ileum development are altered, and an augmented immune response increases rates of offspring mortality. Collectively, we describe a translationally relevant model to examine the defined role of specific human microbial communities on offspring health outcomes, and demonstrate that the prenatal environment dramatically shapes the postnatal response to inoculation.
Collapse
Affiliation(s)
- Eldin Jašarević
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Elizabeth M Hill
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Patrick J Kane
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lindsay Rutt
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Trevonn Gyles
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lillian Folts
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kylie D Rock
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christopher D Howard
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kathleen E Morrison
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jacques Ravel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tracy L Bale
- Center for Epigenetic Research in Child Health and Brain Development, University of Maryland, School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
29
|
Cai J, Cui X, Wang X, You L, Ji C, Cao Y. A Novel Anti-Infective Peptide BCCY-1 With Immunomodulatory Activities. Front Immunol 2021; 12:713960. [PMID: 34367182 PMCID: PMC8339908 DOI: 10.3389/fimmu.2021.713960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 01/05/2023] Open
Abstract
Antibiotic resistance has been considered to be a global threat which underscores the need to develop novel anti-infective therapeutics. Modulation of innate immunity by synthetic peptides is an attractive strategy to overcome this circumstance. We recently reported that BCCY-1, a human β-casein-derived peptide displays regulatory activities on monocytes, thereby enhancing their actions in innate immune responses. However, the function of peptide BCCY-1 in host defense against infection remains unknown. In this study, we investigated the in vivo characteristics and effects of peptide BCCY-1 in mouse models of bacterial infection. Following intraperitoneal injection, the peptide BCCY-1 exhibited high level of cellular uptake by monocytes without obvious toxicities. Results revealed that peptide BCCY-1, but not the scrambled version, stimulated the chemokine production and monocyte recruitment in vivo. Treatment with BCCY-1 enhanced the pathogen clearance and protected mice against lethal infections. Because the anti-infective effects of BCCY-1 was abolished by in vivo depletion of monocytes/macrophages rather than lymphocytes and granulocytes, we conclude that monocytes/macrophages are key effector cells in BCCY-1-mediated anti-infective protection. Additionally, BCCY-1 lacks direct antimicrobial activity. To our knowledge, a human β-casein-derived peptide that counters infection by selective regulation of innate immunity has not been reported previously. These results suggest peptide BCCY-1 as a promising alternative approach and a valuable complement to current anti-infective strategy.
Collapse
Affiliation(s)
- Jinyang Cai
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Lianghui You
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Ren S, Pan X, Hui Y, Kot W, Gao F, Sangild PT, Nguyen DN. Subclinical necrotizing enterocolitis-induced systemic immune suppression in neonatal preterm pigs. Am J Physiol Gastrointest Liver Physiol 2021; 321:G18-G28. [PMID: 34009048 DOI: 10.1152/ajpgi.00408.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Preterm infants are at high risks of sepsis and necrotizing enterocolitis (NEC). Some develop sepsis shortly after suspected or confirmed NEC, implying that NEC may predispose to sepsis but the underlying mechanisms are unknown. Using NEC-sensitive preterm pigs as models, we investigated the immune status in animals following development of subclinical NEC-like lesions with variable severities. Caesarean-delivered preterm pigs were reared until day 5 or day 9. Blood was analyzed for T-cell subsets, neutrophil phagocytosis, transcriptomics, and immune responses to in vitro LPS challenge. Gut tissues were used for histology and cytokine analyses. Pigs with/without macroscopic NEC lesions were scored as healthy, mild, or severe NEC. Overall NEC incidence was similar on day 5 and day 9 (61%-62%) but with lower severity on day 9, implying gradual mucosal repair following the early phase of NEC. Pigs with NEC showed decreased goblet cell density and increased MPO+ and CD3+ cell infiltration in the distal small intestine or colon. Mild or severe NEC lesions had limited effects on circulating parameters on day 5. On day 9, pigs with NEC lesions (especially severe lesions) showed systemic immune suppression, as indicated by elevated Treg frequency, impaired neutrophil phagocytosis, low expression of genes related to innate immunity and Th1 polarization, and diminished LPS-induced immune responses. In conclusion, we shows evidence for NEC-induced systemic immune suppression, even with mild and subclinical NEC lesions. The results help to explain that preterm infants suffering from NEC may show high sensitivity to later secondary infections and sepsis.NEW & NOTEWORTHY Necrotizing enterocolitis (NEC) and sepsis are common diseases in preterm infants. Many develop sepsis following an episode of suspected NEC, suggesting NEC as a predisposing factor for sepsis but mechanisms are unclear. Using preterm pigs as a model, now we show that subclinical NEC lesions, independent of clinical confounding factors, induces systemic immune suppression. The results may help to explain the increased risks of infection and sepsis in preterm infants with previous NEC diagnosis.
Collapse
Affiliation(s)
- Shuqiang Ren
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, People's Republic of China
| | - Xiaoyu Pan
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Yan Hui
- Department of Food Science, University of Copenhagen, Denmark
| | - Witold Kot
- Department of Plant and Environmental Sciences, University of Copenhagen, Denmark
| | - Fei Gao
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, People's Republic of China
| | - Per T Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| |
Collapse
|
31
|
Grzywa TM, Nowis D, Golab J. The role of CD71 + erythroid cells in the regulation of the immune response. Pharmacol Ther 2021; 228:107927. [PMID: 34171326 DOI: 10.1016/j.pharmthera.2021.107927] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Complex regulation of the immune response is necessary to support effective defense of an organism against hostile invaders and to maintain tolerance to harmless microorganisms and autoantigens. Recent studies revealed previously unappreciated roles of CD71+ erythroid cells (CECs) in regulation of the immune response. CECs physiologically reside in the bone marrow where erythropoiesis takes place. Under stress conditions, CECs are enriched in some organs outside of the bone marrow as a result of extramedullary erythropoiesis. However, the role of CECs goes well beyond the production of erythrocytes. In neonates, increased numbers of CECs contribute to their vulnerability to infectious diseases. On the other side, neonatal CECs suppress activation of immune cells in response to abrupt colonization with commensal microorganisms after delivery. CECs are also enriched in the peripheral blood of pregnant women as well as in the placenta and are responsible for the regulation of feto-maternal tolerance. In patients with cancer, anemia leads to increased frequency of CECs in the peripheral blood contributing to diminished antiviral and antibacterial immunity, as well as to accelerated cancer progression. Moreover, recent studies revealed the role of CECs in HIV and SARS-CoV-2 infections. CECs use a full arsenal of mechanisms to regulate immune response. These cells suppress proinflammatory responses of myeloid cells and T-cell proliferation by the depletion of ʟ-arginine by arginase. Moreover, CECs produce reactive oxygen species to decrease T-cell proliferation. CECs also secrete cytokines, including transforming growth factor β (TGF-β), which promotes T-cell differentiation into regulatory T-cells. Here, we comprehensively describe the role of CECs in orchestrating immune response and indicate some therapeutic approaches that might be used to regulate their effector functions in the treatment of human conditions.
Collapse
Affiliation(s)
- Tomasz M Grzywa
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Doctoral School, Medical University of Warsaw, Zwirki and Wigury 61 Street, 02-091 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Centre of Preclinical Research, Medical University of Warsaw, Banacha 1b Street, 02-097 Warsaw, Poland.
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To review recent evidence on the capacity of vitamin D to prevent atopic disease, focussing on food allergy and asthma, and potential underlying mechanisms. RECENT FINDINGS The incidence of allergic disease continues to increase worldwide. Vitamin D status is influenced by sun exposure and dietary intake. Vitamin D deficiency is linked to an increased incidence of allergic disease and asthma. These associations are generally strongest in early life. The capacity of vitamin D to enhance antimicrobial pathways, promote peripheral immunological tolerance and maintain mucosal barrier integrity may underlie these associations. Interventional studies have addressed the capacity of vitamin D supplementation in utero and early life to reduce the incidence of disease. Ancillary studies have provided insights into potential biological mechanisms linked to these effects. SUMMARY Observational studies show an inverse association between vitamin D levels and development of food allergy and asthma. Secondary analyses of two recent interventional studies suggest that achieving vitamin D sufficiency throughout pregnancy reduces the incidence of asthma/recurrent wheeze at 3 years. Longitudinal studies of vitamin D requirements in utero and postnatally, better understanding of factors that influence bioavailability of vitamin D and mechanistic insights into vitamin D effects on neonatal-specific immune pathways are awaited.
Collapse
|
33
|
Brophy-Williams S, Fidanza M, Marchant A, Way S, Kollmann TR. One vaccine for life: Lessons from immune ontogeny. J Paediatr Child Health 2021; 57:782-785. [PMID: 33860973 DOI: 10.1111/jpc.15511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 01/11/2021] [Accepted: 04/05/2021] [Indexed: 11/30/2022]
Abstract
There remains a general misconception that the immune status of the fetus and neonate is immature or insufficient. However, emerging research in immune ontogeny prompts reconsideration of this orthodoxy, reframing this period instead as one of unique opportunity. Vaccine responses (qualitative and quantitative) vary between individuals, and across demographic cohorts. Elements of baseline immune status and function predict vaccine response - some of these factors are well described, others remain a subject of ongoing research, especially with the rapidly expanding field of 'omics' research, enabled by development of highly granular immune profiling techniques and increasing computational capacity. Age is one of the strongest predictive factors associated with variability in the response to vaccination; and predictable variation in response to vaccination is a key to identify the crucial underlying mechanisms. Specifically, circulating maternal antibody in the young infant can modulate immune response to vaccination, acting as an 'undercover adjuvant' that, counter to current dogma, may offer a pathway to longer lasting, higher quality immune response to vaccination. Exciting avenues for novel research in this area have the potential to dramatically alter how we protect the world's most vulnerable population - the very young.
Collapse
Affiliation(s)
- Sam Brophy-Williams
- Department of Infectious Diseases, Perth Childrens Hospital, Child and Adolescent Health Service, Perth, Western Australia, Australia
| | - Mario Fidanza
- Systems Vaccinology, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | - SingSing Way
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital, Cincinnati, Ohio, United States
| | - Tobias R Kollmann
- Systems Vaccinology, Telethon Kids Institute, Perth, Western Australia, Australia
| |
Collapse
|
34
|
Saghafian-Hedengren S, Sverremark-Ekström E, Nilsson A. T Cell Subsets During Early Life and Their Implication in the Treatment of Childhood Acute Lymphoblastic Leukemia. Front Immunol 2021; 12:582539. [PMID: 33763058 PMCID: PMC7982872 DOI: 10.3389/fimmu.2021.582539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The immune system plays a major role in recognizing and eliminating malignant cells, and this has been exploited in the development of immunotherapies aimed at either activating or reactivating the anti-tumor activity of a patient's immune system. A wide range of therapeutic approaches involving T lymphocytes, such as programmed cell death protein ligand-1 (PDL-1) inhibitors, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) blockers, and CD19-targeted T-cell therapy through chimeric antigen receptor (CAR)-T cells or CD19/CD3 bi-specific T-cell engagers, have been introduced to the field of oncology, leading to significant improvements in overall survival of adult cancer patients. During the past few years, the availability and approval of T-cell based immunotherapies have become a reality also for the treatment of childhood cancers. However, the distribution, ratio of regulatory to effector cells and the quality of T-cell responses early in life are distinct from those during adolescence and adulthood, raising the possibility that these differences impact the efficacy of immunotherapy. Herein we provide a brief overview of the properties of conventional T cell subsets during early life. Focusing on the most common cancer type during childhood, acute lymphoblastic leukemia (ALL), we describe how current conventional therapies used against ALL influence the T-cell compartment of small children. We describe early life T-cell responses in relation to immunotherapies engaging T-cell anticancer reactivity and present our opinion that it is not only immaturity of the adaptive immune system, but also the impact of an immunosuppressive environment that may prove disadvantageous in the setting of immunotherapies targeting pediatric cancer cells.
Collapse
Affiliation(s)
- Shanie Saghafian-Hedengren
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna Nilsson
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
The Influence of Immune Immaturity on Outcome After Virus Infections. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:641-650. [PMID: 33551039 PMCID: PMC8042246 DOI: 10.1016/j.jaip.2020.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
Maturation of the adaptive immune response is typically thought to improve outcome to virus infections. However, long-standing observations of natural infections with old viruses such as Epstein-Barr virus and newer observations of emerging viruses such as severe acute respiratory syndrome coronavirus 2 responsible for COVID-19 suggest that immune immaturity may be beneficial for outcome. Mechanistic studies and studies of patients with inborn errors of immunity have revealed that immune dysregulation reflecting inappropriate antibody and T-cell responses plays a crucial role in causing bystander inflammation and more severe disease. Further evidence supports a role for innate immunity in normally regulating adaptive immune responses. Thus, changes in immune responses that normally occur with age may help explain an apparent protective role of immune immaturity during virus infections.
Collapse
|
36
|
Wu D, Ding Y, Yao K, Gao W, Wang Y. Antimicrobial Resistance Analysis of Clinical Escherichia coli Isolates in Neonatal Ward. Front Pediatr 2021; 9:670470. [PMID: 34113589 PMCID: PMC8185016 DOI: 10.3389/fped.2021.670470] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/26/2021] [Indexed: 11/26/2022] Open
Abstract
Background: Escherichia coli (E. coli) column for one of the most common pathogens causing neonatal infections. The emergence of antibiotic-resistant bacteria is a major cause of treatment failure in infected newborns. The purpose of this study was to describe antibiotic and multidrug resistance of E. coli strains isolated from neonates with infection throughout the years 2009-2011. Methods: The antimicrobial susceptibility testing of E. coli strains to selected antibiotics was assessed using the E-test technique on the Mueller-Hinton agar. The antimicrobial tests included ceftazidime, cefuroxime, cefatriaxone, amoxicillin, amoxicillin-clavulanic acid, cefoperazone- sulbactam, meropenem, gentamicin, ciprofloxacin, and sulfonamides. Results: A total of 100 E. coli strains were isolated from sputum (n = 78), blood (n = 10), cerebrospinal fluid (n = 5), and umbilical discharge (n = 7) samples of hospitalized neonates at the Beijing Children's Hospital. The highest rate of E. coli resistance was found in amoxicillin (85%), followed by cefuroxime (65%), and cefatriaxone (60%), respectively. A total of 6 and 5% of all isolates were only resistant to amoxicillin/clavulanic acid and cefoperazone -sulbactam. The rates of resistance to ceftazidime, gentamicin, ciprofloxacin, and sulfonamides were 31, 20, 33, and 47%, respectively. All isolates were susceptible to meropenem. Approximately 26% of all E. coli isolates were multidrug-resistant. The detection rate of ESBL-Producing E. coli was 55%. Conclusions: Multi-drug-resistant E. coli has become an important and complex problem in clinical treatment, and it is thus essential to monitor E. coli resistance in neonates.
Collapse
Affiliation(s)
- Dan Wu
- Department of Neonatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yijun Ding
- Department of Neonatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Kaihu Yao
- Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Gao
- Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yajuan Wang
- Department of Neonatology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
37
|
Elahi S, Vega-López MA, Herman-Miguel V, Ramírez-Estudillo C, Mancilla-Ramírez J, Motyka B, West L, Oyegbami O. CD71 + Erythroid Cells in Human Neonates Exhibit Immunosuppressive Properties and Compromise Immune Response Against Systemic Infection in Neonatal Mice. Front Immunol 2020; 11:597433. [PMID: 33329589 PMCID: PMC7732591 DOI: 10.3389/fimmu.2020.597433] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/29/2020] [Indexed: 01/08/2023] Open
Abstract
Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+ erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.
Collapse
Affiliation(s)
- Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB, Canada.,Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Marco Antonio Vega-López
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Vladimir Herman-Miguel
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Carmen Ramírez-Estudillo
- Dept. Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Javier Mancilla-Ramírez
- Escuela Superior deMedicina, Instituto Politecnico Nacional, Hospital de la Mujer, Secretaria de Salud, Mexico City, Mexico
| | - Bruce Motyka
- Alberta Transplant Institute and the Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Lori West
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton AB, Canada.,Alberta Transplant Institute and the Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - Olaide Oyegbami
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Elahi S. Neonatal and Children’s Immune System and COVID-19: Biased Immune Tolerance versus Resistance Strategy. THE JOURNAL OF IMMUNOLOGY 2020; 205:1990-1997. [DOI: 10.4049/jimmunol.2000710] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The recent outbreak of COVID-19 has emerged as a major global health concern. Although susceptible to infection, recent evidence indicates mostly asymptomatic or mild presentation of the disease in infants, children, and adolescents. Similar observations were made for acute respiratory infections caused by other coronaviruses (severe acute respiratory syndrome and Middle East respiratory syndrome). These observations suggest that the immune system behaves differently in children than adults. Recent developments in the field demonstrated fundamental differences in the neonatal immune system as compared with adults, whereby infants respond to microorganisms through biased immune tolerance rather than resistance strategies. Similarly, more frequent/recent vaccinations in children and younger populations may result in trained immunity. Therefore, the physiological abundance of certain immunosuppressive cells, a tightly regulated immune system, and/or exposure to attenuated vaccines may enhance trained immunity to limit excessive immune reaction to COVID-19 in the young.
Collapse
Affiliation(s)
- Shokrollah Elahi
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G2E1, Canada
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G1Z2, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G2E1, Canada; and
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta T6G2E1, Canada
| |
Collapse
|
39
|
Roodsant T, Navis M, Aknouch I, Renes IB, van Elburg RM, Pajkrt D, Wolthers KC, Schultsz C, van der Ark KCH, Sridhar A, Muncan V. A Human 2D Primary Organoid-Derived Epithelial Monolayer Model to Study Host-Pathogen Interaction in the Small Intestine. Front Cell Infect Microbiol 2020; 10:272. [PMID: 32656095 PMCID: PMC7326037 DOI: 10.3389/fcimb.2020.00272] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Gut organoids are stem cell derived 3D models of the intestinal epithelium that are useful for studying interactions between enteric pathogens and their host. While the organoid model has been used for both bacterial and viral infections, this is a closed system with the luminal side being inaccessible without microinjection or disruption of the organoid polarization. In order to overcome this and simplify their applicability for transepithelial studies, permeable membrane based monolayer approaches are needed. In this paper, we demonstrate a method for generating a monolayer model of the human fetal intestinal polarized epithelium that is fully characterized and validated. Proximal and distal small intestinal organoids were used to generate 2D monolayer cultures, which were characterized with respect to epithelial cell types, polarization, barrier function, and gene expression. In addition, viral replication and bacterial translocation after apical infection with enteric pathogens Enterovirus A71 and Listeria monocytogenes were evaluated, with subsequent monitoring of the pro-inflammatory host response. This human 2D fetal intestinal monolayer model will be a valuable tool to study host-pathogen interactions and potentially reduce the use of animals in research.
Collapse
Affiliation(s)
- Thomas Roodsant
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Marit Navis
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Ikrame Aknouch
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Viroclinics Xplore, Schaijk, Netherlands
| | - Ingrid B Renes
- Danone Nutricia Research, Utrecht, Netherlands.,Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Ruurd M van Elburg
- Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Katja C Wolthers
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Constance Schultsz
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Kees C H van der Ark
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Adithya Sridhar
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Vanesa Muncan
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
40
|
Ng S, Strunk T, Lee AH, Gill EE, Falsafi R, Woodman T, Hibbert J, Hancock REW, Currie A. Whole blood transcriptional responses of very preterm infants during late-onset sepsis. PLoS One 2020; 15:e0233841. [PMID: 32479514 PMCID: PMC7263612 DOI: 10.1371/journal.pone.0233841] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Host immune responses during late-onset sepsis (LOS) in very preterm infants are poorly characterised due to a complex and dynamic pathophysiology and challenges in working with small available blood volumes. We present here an unbiased transcriptomic analysis of whole peripheral blood from very preterm infants at the time of LOS. METHODS RNA-Seq was performed on peripheral blood samples (6-29 days postnatal age) taken at the time of suspected LOS from very preterm infants <30 weeks gestational age. Infants were classified based on blood culture positivity and elevated C-reactive protein concentrations as having confirmed LOS (n = 5), possible LOS (n = 4) or no LOS (n = 9). Bioinformatics and statistical analyses performed included pathway over-representation and protein-protein interaction network analyses. Plasma cytokine immunoassays were performed to validate differentially expressed cytokine pathways. RESULTS The blood leukocyte transcriptional responses of infants with confirmed LOS differed significantly from infants without LOS (1,317 differentially expressed genes). However, infants with possible LOS could not be distinguished from infants with no LOS or confirmed LOS. Transcriptional alterations associated with LOS included genes involved in pathogen recognition (mainly TLR pathways), cytokine signalling (both pro-inflammatory and inhibitory responses), immune and haematological regulation (including cell death pathways), and metabolism (altered cholesterol biosynthesis). At the transcriptional-level cytokine responses during LOS were characterised by over-representation of IFN-α/β, IFN-γ, IL-1 and IL-6 signalling pathways and up-regulation of genes for inflammatory responses. Infants with confirmed LOS had significantly higher levels of IL-1α and IL-6 in their plasma. CONCLUSIONS Blood responses in very preterm infants with LOS are characterised by altered host immune responses that appear to reflect unbalanced immuno-metabolic homeostasis.
Collapse
Affiliation(s)
- Sherrianne Ng
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Division of the Institute of Reproductive and Developmental Biology, Imperial College Parturition Research Group, Imperial College London, London, United Kingdom
- March of Dimes European Prematurity Research Centre, Imperial College London, London, United Kingdom
| | - Tobias Strunk
- Department of Health, Neonatal Directorate, King Edward Memorial Hospital, Child and Adolescent Health Service, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Amy H. Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin E. Gill
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tabitha Woodman
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Julie Hibbert
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Robert E. W. Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Currie
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
- * E-mail:
| |
Collapse
|
41
|
Michels KR, Lambrecht NJ, Carson WF, Schaller MA, Lukacs NW, Bermick JR. The Role of Iron in the Susceptibility of Neonatal Mice to Escherichia coli K1 Sepsis. J Infect Dis 2020; 220:1219-1229. [PMID: 31136646 DOI: 10.1093/infdis/jiz282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/24/2019] [Indexed: 12/23/2022] Open
Abstract
Sepsis from Escherichia coli expressing the K1 antigen is a leading cause of death in neonates. In a murine model, E. coli K1 grew rapidly in the peritoneal cavity of neonatal mice, causing fatal disease. In contrast, adult mice cleared the infection. Neonatal mice mounted a rapid and equivalent antimicrobial immune response compared to adult mice. Interestingly, peritoneal fluid from neonatal mice contained significantly more total iron than that of adult mice, which was sufficient to support enhanced E. coli growth. Transient iron overload in adult mice infected with E. coli resulted in 100% mortality. Maternal diet-induced mild iron deficiency decreased offspring peritoneal iron, decreased bacterial growth, and conferred protection against sepsis. Taken together, neonatal susceptibility to E. coli K1 sepsis is enhanced by a localized excess of peritoneal iron that allows for unchecked bacterial growth. Targeting this excess iron may provide a new therapeutic target in human patients.
Collapse
Affiliation(s)
- Kathryn R Michels
- Department of Pathology, Michigan Medicine, School of Public Health, University of Michigan, Ann Arbor
| | - Nathalie J Lambrecht
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor
| | - William F Carson
- Department of Pathology, Michigan Medicine, School of Public Health, University of Michigan, Ann Arbor
| | - Matthew A Schaller
- Department of Pathology, Michigan Medicine, School of Public Health, University of Michigan, Ann Arbor
| | - Nicholas W Lukacs
- Department of Pathology, Michigan Medicine, School of Public Health, University of Michigan, Ann Arbor.,Mary H. Weiser Food Allergy Center, Department of Pediatrics, Michigan Medicine, Ann Arbor
| | - Jennifer R Bermick
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, Ann Arbor
| |
Collapse
|
42
|
Long-Term Incidence of Infectious-Related Hospitalizations of Offspring Born to Mothers With Intrauterine Device: A Retrospective Population-Based Cohort Study. Pediatr Infect Dis J 2020; 39:325-329. [PMID: 31929381 DOI: 10.1097/inf.0000000000002575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pregnancy with an Intrauterine device (IUD) is a risk factor for adverse pregnancy outcomes. The purpose of this study was to examine a possible association between presence of IUD during pregnancy and long-term incidence of infectious-related hospitalizations of the offspring. METHODS A population-based cohort analysis was performed including all singleton deliveries between 1991 and 2014 at a single tertiary hospital. Primary exposure was defined as delivery of an infant to a mother who conceived with an IUD, that was either removed or retained during pregnancy. Offspring of mothers who conceived without an IUD comprised the comparison group. The main outcome evaluated was infectious-related hospitalizations of the offspring up to the age of 18 years. RESULTS The number of deliveries that met the inclusion criteria was 227,431, of which 209 were to mothers who conceived with an IUD that was removed and 52 were to mothers who retained their device. Long-term incidence of infections-related hospitalizations was compared between the groups (12.5% in the retained IUD, 12.9% in the removed IUD and 11.2% in the No-IUD group, P = 0.638). A Kaplan-Meier survival curve did not demonstrate a significantly higher cumulative incidence of infectious-related hospitalizations in offspring of women who conceived with an IUD (log-rank P = 0.340). In a Cox regression model, while controlling for confounders such as maternal age and preterm birth, conceiving with an IUD was not found to be independently associated with long-term incidence of infectious-related hospitalizations of the offspring. CONCLUSION Offspring of women conceiving with an IUD are not at increased risk for long-term incidence of infectious-related hospitalizations.
Collapse
|
43
|
Tsafaras GP, Ntontsi P, Xanthou G. Advantages and Limitations of the Neonatal Immune System. Front Pediatr 2020; 8:5. [PMID: 32047730 PMCID: PMC6997472 DOI: 10.3389/fped.2020.00005] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022] Open
Abstract
During early post-natal life, neonates must adjust to the transition from the sheltered intra-uterine environment to the microbe-laden external world, wherein they encounter a constellation of antigens and the colonization by the microbiome. At this vulnerable stage, neonatal immune responses are considered immature and present significant differences to those of adults. Pertinent to innate immunity, functional and quantitative deficiencies in antigen-presenting cells and phagocytes are often documented. Exposure to environmental antigens and microbial colonization is associated with epigenetic immune cell reprogramming and activation of effector and regulatory mechanisms that ensure age-depended immune system maturation and prevention of tissue damage. Moreover, neonatal innate immune memory has emerged as a critical mechanism providing protection against infectious agents. Still, in neonates, inexperience to antigenic exposure, along with enhancement of tissue-protective immunosuppressive mechanisms are often associated with severe immunopathological conditions, including sepsis and neurodevelopmental disorders. Despite significant advances in the field, adequate vaccination in newborns is still in its infancy due to elemental restrictions associated also with defective immune responses. In this review, we provide an overview of neonatal innate immune cells, highlighting phenotypic and functional disparities with their adult counterparts. We also discuss the effects of epigenetic modifications and microbial colonization on the regulation of neonatal immunity. A recent update on mechanisms underlying dysregulated neonatal innate immunity and linked infectious and neurodevelopmental diseases is provided. Understanding of the mechanisms that augment innate immune responsiveness in neonates may facilitate the development of improved vaccination protocols that can protect against pathogens and organ damage.
Collapse
Affiliation(s)
- George P Tsafaras
- Cellular Immunology Lab, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Polyxeni Ntontsi
- Second Respiratory Medicine Department, 'Attikon' University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Georgina Xanthou
- Cellular Immunology Lab, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
44
|
Ren S, Pan X, Gao F, Sangild PT, Nguyen DN. Prenatal inflammation suppresses blood Th1 polarization and gene clusters related to cellular energy metabolism in preterm newborns. FASEB J 2019; 34:2896-2911. [PMID: 31908027 DOI: 10.1096/fj.201902629r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022]
Abstract
Chorioamnionitis (CA, fetal membrane inflammation) predisposes to preterm birth and is associated with increased neonatal infection risk, but the separate effects of prematurity, CA, and postnatal adaptations on this risk are unclear. Using pigs as models for infants, we examined the systemic immune-metabolic status in cesarean-delivered preterm pigs, with and without CA induced by intra-amniotic (IA) LPS exposure. At birth, cord blood of preterm pigs showed neutropenia and low expressions of innate and adaptive immune genes, relative to term pigs. IA LPS induced CA and fetal systemic innate immune activation via complement and neutrophil-related pathways. These were mainly modulated via cellular regulations rather than granulopoiesis, as validated by the in vitro LPS stimulation of cord blood. After birth, IA LPS-exposed preterm pigs did not follow normal immune-metabolic ontogenies found in fetuses or newborns without prenatal insults, but showed consistently high levels of Treg, impaired Th1 polarization, and reduced expressions of multiple genes related to cellular oxidative phosphorylation and ribosomal activities. In conclusion, our results provide cellular and molecular evidence for CA-induced distinct neonatal immune-metabolic status with increased disease tolerance strategy, suggesting mechanisms for the clinical observation of elevated sepsis risks in immune-compromised preterm infants born with CA.
Collapse
Affiliation(s)
- Shuqiang Ren
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Xiaoyu Pan
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Fei Gao
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Per T Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.,Hans Christian Andersen Children's Hospital, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
45
|
Brook B, Harbeson D, Amenyogbe N, Ben-Othman R, Kollmann TR, Aniba R. Robust health-score based survival prediction for a neonatal mouse model of polymicrobial sepsis. PLoS One 2019; 14:e0218714. [PMID: 31233529 PMCID: PMC6590826 DOI: 10.1371/journal.pone.0218714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/09/2019] [Indexed: 01/30/2023] Open
Abstract
Infectious disease and sepsis represent a serious problem for all, but especially in early life. Much of the increase in morbidity and mortality due to infection in early life is presumed to relate to fundamental differences between neonatal and adult immunity. Mechanistic insight into the way newborns' immune systems handle infectious threats is lacking; as a result, there has only been limited success in providing effective immunomodulatory interventions to reduce infectious mortality. Given the complexity of the host-pathogen interactions, neonatal mouse models can offer potential avenues providing valuable data. However, the small size of neonatal mice hampers the ability to collect biological samples without sacrificing the animals. Further, the lack of a standardized metric to quantify newborn mouse health increases reliance on correlative biomarkers without a known relationship to 'clinical' outcome. To address this bottleneck, we developed a system that allows assessment of neonatal mouse health in a readily standardized and quantifiable manner. The resulting health scores require no special equipment or sample collection and can be assigned in less than 20 seconds. Importantly, the health scores are highly predictive of survival. A classifier built on our health score revealed a positive relationship between reduced bacterial load and survival, demonstrating how this scoring system can be used to bridge the gap between assumed relevance of biomarkers and the clinical outcome of interest. Adoption of this scoring system will not only provide a robust metric to assess health of newborn mice but will also allow for objective, prospective studies of infectious disease and possible interventions in early life.
Collapse
Affiliation(s)
- Byron Brook
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| | - Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Radhouane Aniba
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Lakhdari O, Yamamura A, Hernandez GE, Anderson KK, Lund SJ, Oppong-Nonterah GO, Hoffman HM, Prince LS. Differential Immune Activation in Fetal Macrophage Populations. Sci Rep 2019; 9:7677. [PMID: 31118442 PMCID: PMC6531440 DOI: 10.1038/s41598-019-44181-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/02/2019] [Indexed: 02/06/2023] Open
Abstract
Distinct macrophage subsets populate the developing embryo and fetus in distinct waves. However little is known about the functional differences between in utero macrophage populations or how they might contribute to fetal and neonatal immunity. Here we tested the innate immune response of mouse macrophages derived from the embryonic yolk sac and from fetal liver. When isolated from liver or lung, CD11bHI fetal liver derived macrophages responded to the TLR4 agonist LPS by expressing and releasing inflammatory cytokines. However F4/80HI macrophages from the yolk sac did not respond to LPS treatment. While differences in TLR4 expression did not appear to explain these data, F4/80HI macrophages had much lower NLRP3 inflammasome expression compared to CD11bHI macrophages. Gene expression profiling also demonstrated LPS-induced expression of inflammatory genes in CD11bHI macrophages, but not in F4/80HI cells. Genes expressed in LPS-treated CD11bHI macrophages were more likely to contain predicted NF-κB binding sites in their promoter regions. Our data show that CD11bHI macrophages derived from fetal liver are the major pro-inflammatory cells in the developing fetus. These findings could have important implications in better understanding the fetal inflammatory response and the unique features of neonatal immunity.
Collapse
Affiliation(s)
- Omar Lakhdari
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Asami Yamamura
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Gilberto E Hernandez
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Kathryn K Anderson
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Sean J Lund
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Gertrude O Oppong-Nonterah
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Hal M Hoffman
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Lawrence S Prince
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA.
| |
Collapse
|
47
|
Meng X, Dunsmore G, Koleva P, Elloumi Y, Wu RY, Sutton RT, Ambrosio L, Hotte N, Nguyen V, Madsen KL, Dieleman LA, Chen H, Huang V, Elahi S. The Profile of Human Milk Metabolome, Cytokines, and Antibodies in Inflammatory Bowel Diseases Versus Healthy Mothers, and Potential Impact on the Newborn. J Crohns Colitis 2019; 13:431-441. [PMID: 30418545 PMCID: PMC6441305 DOI: 10.1093/ecco-jcc/jjy186] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS For women with inflammatory bowel disease [IBD], it is not very well known how IBD or IBD treatment affects their breast milk components. We aimed to investigate whether breast milk composition differs in healthy control [HC] versus IBD mothers in terms of antibodies, cytokines, and metabolite,s to identify potential impact of IBD breast milk on neonatal immune system. METHODS Breast milk specimens from HC [n = 17] and IBD [n = 31 for Crohn's disease [CD]; and n = 41 for ulcerative colitis [UC]; were collected at 3 and 6 months postpartum [PP3] and [PP6], respectively. Faecal samples were also collected. Cytokines and immunoglobulins [IgA/IgG/IgE] were analysed by multiplex Meso Scale Discovery [MSD] and commercial kits. Moreover, breast milk metabolites were analysed by 1H nuclear magnetic resonance [NMR]. RESULTS We found that breast milk from IBD mothers showed significantly lower levels of IgA, sugar metabolite [lactose], and 2-aminobutyrate. In contrast, we observed that breast milk from mothers with IBD had increased levels of pro-inflammatory cytokines and higher energy metabolites [lactate and succinate] than milk from healthy mothers. In addition, we noticed that the type of treatment [5-aminosalicylic acid versus biologics] influenced the milk cytokines and metabolites profile. CONCLUSIONS The reduction in immunoprotective components of IBD breast milk such as sIgA and lactose theoretically may modulate the potential protective effects of breastfeeding. On the other hand, presence of higher levels of pro-inflammatory cytokines, lactate, and succinate may predispose the offspring to an inflammatory condition or impact on the gut microbiome. Better understanding of the role of succinate in infants and its potential effects on microbiome or mucosal immunity merits further investigations.
Collapse
Affiliation(s)
- Xuanyi Meng
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada,State Key Laboratory of Food Science and Technology, Nanchang University. Nanchang, China
| | - Garett Dunsmore
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Petya Koleva
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yesmine Elloumi
- Division of Gastroenterology, University of Alberta, AB, Canada
| | - Richard You Wu
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Lindsy Ambrosio
- Division of Gastroenterology, University of Alberta, AB, Canada
| | - Naomi Hotte
- Division of Gastroenterology, University of Alberta, AB, Canada
| | - Vivian Nguyen
- Division of Gastroenterology, University of Alberta, AB, Canada
| | - Karen L Madsen
- Division of Gastroenterology, University of Alberta, AB, Canada
| | | | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University. Nanchang, China
| | - Vivian Huang
- Division of Gastroenterology, University of Alberta, AB, Canada,Division of Gastroenterology, University of Toronto, Mount Sinai Hospital, Toronto, ON, Canada
| | - Shokrollah Elahi
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada,Corresponding author: Shokrollah Elahi, PhD, 7020L, Katz Group Centre for Pharmacology and Health Research, 11361-87th Ave NW, Edmonton, AB, T6G2E1, Canada. Tel.: 780-492-1336; fax: 780-492-7466;
| |
Collapse
|
48
|
Pietrasanta C, Pugni L, Ronchi A, Bottino I, Ghirardi B, Sanchez-Schmitz G, Borriello F, Mosca F, Levy O. Vascular Endothelium in Neonatal Sepsis: Basic Mechanisms and Translational Opportunities. Front Pediatr 2019; 7:340. [PMID: 31456998 PMCID: PMC6700367 DOI: 10.3389/fped.2019.00340] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
Neonatal sepsis remains a major health issue worldwide, especially for low-birth weight and premature infants, with a high risk of death and devastating sequelae. Apart from antibiotics and supportive care, there is an unmet need for adjunctive treatments to improve the outcomes of neonatal sepsis. Strong and long-standing research on adult patients has shown that vascular endothelium is a key player in the pathophysiology of sepsis and sepsis-associated organ failure, through a direct interaction with pathogens, leukocytes, platelets, and the effect of soluble circulating mediators, in part produced by endothelial cells themselves. Despite abundant evidence that the neonatal immune response to sepsis is distinct from that of adults, comparable knowledge on neonatal vascular endothelium is much more limited. Neonatal endothelial cells express lower amounts of adhesion molecules compared to adult ones, and present a reduced capacity to neutralize reactive oxygen species. Conversely, available evidence on biomarkers of endothelial damage in neonates is not as robust as in adult patients, and endothelium-targeted therapeutic opportunities for neonatal sepsis are almost unexplored. Here, we summarize current knowledge on the structure of neonatal vascular endothelium, its interactions with neonatal immune system and possible endothelium-targeted diagnostic and therapeutic tools for neonatal sepsis. Furthermore, we outline areas of basic and translational research worthy of further study, to shed light on the role of vascular endothelium in the context of neonatal sepsis.
Collapse
Affiliation(s)
- Carlo Pietrasanta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Lorenza Pugni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Andrea Ronchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Ilaria Bottino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Beatrice Ghirardi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organisation Center of Excellence, Naples, Italy
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
49
|
Harbeson D, Francis F, Bao W, Amenyogbe NA, Kollmann TR. Energy Demands of Early Life Drive a Disease Tolerant Phenotype and Dictate Outcome in Neonatal Bacterial Sepsis. Front Immunol 2018; 9:1918. [PMID: 30190719 PMCID: PMC6115499 DOI: 10.3389/fimmu.2018.01918] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/03/2018] [Indexed: 12/16/2022] Open
Abstract
Bacterial sepsis is one of the leading causes of death in newborns. In the face of growing antibiotic resistance, it is crucial to understand the pathology behind the disease in order to develop effective interventions. Neonatal susceptibility to sepsis can no longer be attributed to simple immune immaturity in the face of mounting evidence that the neonatal immune system is tightly regulated and well controlled. The neonatal immune response is consistent with a "disease tolerance" defense strategy (minimizing harm from immunopathology) whereas adults tend toward a "disease resistance" strategy (minimizing harm from pathogens). One major advantage of disease tolerance is that is less energetically demanding than disease resistance, consistent with the energetic limitations of early life. Immune effector cells enacting disease resistance responses switch to aerobic glycolysis upon TLR stimulation and require steady glycolytic flux to maintain the inflammatory phenotype. Rapid and intense upregulation of glucose uptake by immune cells necessitates an increased reliance on fatty acid metabolism to (a) fuel vital tissue function and (b) produce immunoregulatory intermediates which help control the magnitude of inflammation. Increasing disease resistance requires more energy: while adults have fat and protein stores to catabolize, neonates must reallocate resources away from critical growth and development. This understanding of sepsis pathology helps to explain many of the differences between neonatal and adult immune responses. Taking into account the central role of metabolism in the host response to infection and the severe metabolic demands of early life, it emerges that the striking clinical susceptibility to bacterial infection of the newborn is at its core a problem of metabolism. The evidence supporting this novel hypothesis, which has profound implications for interventions, is presented in this review.
Collapse
Affiliation(s)
- Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Freddy Francis
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Winnie Bao
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nelly A. Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|