1
|
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]
|
2
|
Perveen K, Ferrante A. Protein Kinase C Isozyme Immaturity/Deficiency in Cord Blood Monocytes and Neutrophils. Int J Mol Sci 2024; 25:11665. [PMID: 39519215 PMCID: PMC11546585 DOI: 10.3390/ijms252111665] [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: 09/27/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Reduced/deficient expression of Protein Kinase C (PKC)ζ in Cord blood (CB) T cells is associated with allergy development in children and a propensity to maintain an immature T-helper (Th)2 cytokine profile. In addition, other PKC isozymes are also low in CBTCs. Since previous studies have reported that cord blood/neonatal monocyte and neutrophil functions are significantly lower than cells from adults, it was of interest to see if the CBTC PKC levels were reflected in CB monocytes and neutrophils. Compared to adult blood, CB expresses low levels of PKCα, β2, ε, θ, μ, ζ and λ/ι in monocytes and PKCα, β2, η, θ, μ, ζ and λ/ι in neutrophils. The T-cell PKCζ levels were positively correlated with levels in CB monocytes but not in neutrophils. However, neither the monocytes nor the neutrophil PKCζ were associated with T-cell development towards a Th1 or Th2 cytokine propensity, based on the production of interferon-gamma and interleukin-4 in response to phytohemagglutinin and phorbol myristate acetate. The results demonstrate that some newborn babies display a deficiency in PKC isozymes in monocytes and neutrophils, as reported for T cells. However, unlike T cells, the PKCζ levels of the phagocytes did not correlate with regulation of development towards a Th1 or Th2 cytokine phenotype.
Collapse
Affiliation(s)
- Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia;
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia;
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
3
|
Sedney CJ, Masters J, Callender M, Dewan K, Caulfield A, Harvill ET. Neonatal Neutrophil-mediated Control of Bordetella pertussis Is Disrupted by Pertussis Toxin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:ji2400363. [PMID: 39475256 PMCID: PMC11605672 DOI: 10.4049/jimmunol.2400363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/08/2024] [Indexed: 12/01/2024]
Abstract
The increased susceptibility of infants and young children to some diseases has often been explained as the neonatal immune system (NIS) being incomplete and/or underdeveloped. However, our recent work demonstrated that neonatal mice could clear a Bordetella pertussis (Bp) strain lacking pertussis toxin (PTx) (BpΔptx) much more efficiently than adult mice, indicating that the NIS can be extremely effective, but this ability is highly sensitive to being blocked by PTx. In this article, we investigated immunological mechanisms by which neonates efficiently and rapidly clear BpΔptx to better understand how the NIS functions and how PTx disrupts it. Depleting neutrophils, or blocking their recruitment, inhibited pups' ability to rapidly clear BpΔptx, revealing a critical role for neutrophils. Pups deficient in complement (C3-/-) failed to recruit neutrophils and did not efficiently clear BpΔptx but recovered these abilities upon treatment with C3a. Neutrophil depletion in C3-/- pups led to further failure to control BpΔptx, suggesting that neutrophils and complement have independent roles in rapid clearance of BpΔptx. Depleting or disrupting neutrophils and complement had negligible effect on the rapid growth of wild-type Bp, indicating that PTx blocks these otherwise highly effective aspects of the NIS.
Collapse
Affiliation(s)
- Colleen J. Sedney
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Jillian Masters
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Maiya Callender
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Kalyan Dewan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Amanda Caulfield
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| |
Collapse
|
4
|
da Silveira BP, Cohen ND, Lawhon SD, Watson RO, Bordin AI. Protective immune response against Rhodococcus equi: An innate immunity-focused review. Equine Vet J 2024. [PMID: 39258739 DOI: 10.1111/evj.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024]
Abstract
Rhodococcus equi causes pyogranulomatous pneumonia in foals and immunocompromised people. Despite decades of research efforts, no vaccine is available against this common cause of disease and death in foals. The purpose of this narrative review is to summarise the current understanding of interactions between R. equi and the host innate immune system, to describe features of the immune response that are associated with resistance or susceptibility to R. equi infection, and help guide strategies for developing novel approaches for preventing R. equi infections. Virulence of R. equi in foals has been attributed to the virulence associated protein A which allows intracellular survival in macrophages by preventing acidification of R. equi-containing vacuole. Additionally, foal susceptibility to R. equi infection is associated with immaturity and naivety of innate and adaptive immune systems, while adult horses with fully functional immune system are resistant to pneumonia. Specific interaction between R. equi and innate immune cells can result in bacterial survival or death; learning how to manipulate these responses to control infection is critical to prevent pneumonia in foals. Administration of live vaccines and stimulation of innate immune responses appears to improve foals' immune response and has the potential to overcome the challenges of foal active vaccination and elicit protection against pneumonia.
Collapse
Affiliation(s)
- Bibiana Petri da Silveira
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Noah D Cohen
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Sara D Lawhon
- Department of Veterinary Pathobiology, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Robert O Watson
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, School of Medicine, College Station, Texas, USA
| | - Angela I Bordin
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| |
Collapse
|
5
|
Jung KJ, Cho J, Yang MJ, Hwang JH, Song J. Exposure to polyhexamethyleneguanidine phosphate in early life dampens pulmonary damage compared to adult mice. Chem Biol Interact 2024; 399:111134. [PMID: 38969276 DOI: 10.1016/j.cbi.2024.111134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Polyhexamethyleneguanidine phosphate (PHMG-P) is a biocide of guanidine family that can cause a fatal lung damage if exposed directly to the lungs. No reports exist regarding the toxicity of PHMG-P in neonatal animals. Therefore, this study aimed to determine PHMG-P toxicity in neonatal and 8-week-old mice after they were intranasally instilled with 1.5 mg/kg, 3 mg/kg, and 4.5 mg/kg PHMG-P. PHMG-P lung exposure resulted in more severe pulmonary toxicity in adult mice than in newborn mice. In the high-dose group of newborn mice, a minimal degree of inflammatory cell infiltration and fibrosis in the lung were detected, whereas more severe pathological lesions including granulomatous inflammation, fibrosis, and degeneration of the bronchiolar epithelium were observed in adult mice. At day 4, C-C motif chemokine ligand 2 (CCL2), a potent chemokine for monocytes, was upregulated but recovered to normal levels at day 15 in newborn mice. However, increased CCL2 and IL-6 levels were sustained at day 15 in adult mice. When comparing the differentially expressed genes of newborn and adult mice through RNA-seq analysis, there were expression changes in several genes associated with inflammation in neonates that were similar or different from those in adults. Although no significant lung damage occurred in newborns, growth inhibition was observed which was not reversed until the end of the experiment. Further research is needed to determine how growth inhibition from neonatal exposure to PHMG-P affects adolescent and young adult health.
Collapse
Affiliation(s)
- Kyung Jin Jung
- Immunotoxicology Research Group, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Jeonghee Cho
- Center for Vascular Research, Institute for Basci Science, Daejeon, 34126, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Pathology Research Group, Korea Institute of Toxicology, Jeonbuk, 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
|
6
|
Tung NWH, Edmondson C, Westrupp N, Rosenthal M, Davies JC. Neutrophil-to-lymphocyte ratio as a biomarker of acute pulmonary exacerbations in children with cystic fibrosis: a retrospective cohort study. Arch Dis Child 2024; 109:748-754. [PMID: 38876505 DOI: 10.1136/archdischild-2024-327056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Neutrophils are key contributors to chronic airway inflammation in cystic fibrosis (CF) lung disease, although airway and blood-based neutrophil markers are seldom used. The neutrophil-to-lymphocyte ratio (NLR) is an accessible biomarker, the clinical utility of which has not been adequately studied. OBJECTIVE This study aimed to investigate the characteristics of the NLR in children with CF and its correlations with acute pulmonary exacerbations and spirometry. DESIGN A previous study had collected clinical data from children with CF for a 3-year period between 2016 and 2021. Retrospectively, NLR values were categorised according to patients' clinical status during blood sample collection as 'stable', 'acute pulmonary exacerbation' or 'elective admission for chronic clinical concern'. MAIN OUTCOME MEASURES Demographic characteristics associated with the NLR; changes in NLR values in relation to clinical status; relationship between NLR and lung function. RESULTS 141 children with CF were included. NLR values during clinical stability were higher in females and increased with age. For children admitted for intravenous antibiotics, NLR values significantly increased from clinical stability (median (IQR)=1.13 (0.75-1.51)) to acute pulmonary exacerbations (median (IQR)=1.50 (0.96-2.65), p=0.001), but similar changes were not observed in elective admissions. The NLR was not associated with lung function. CONCLUSIONS The NLR demonstrated associations with clinical status in children with CF with significant elevations during acute pulmonary exacerbations. While its utility as a single-marker measure is limited, monitoring the NLR over time may help identify periods of increased inflammation.
Collapse
Affiliation(s)
| | - Claire Edmondson
- Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole Westrupp
- Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mark Rosenthal
- Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jane C Davies
- Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
7
|
Mincham KT, Lauzon-Joset JF, Read JF, Holt PG, Stumbles PA, Strickland DH. Mapping Lung Hematopoietic Progenitors: Developmental Kinetics and Response to Influenza A Infection. Am J Respir Cell Mol Biol 2024; 71:219-228. [PMID: 38669465 DOI: 10.1165/rcmb.2023-0395oc] [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: 11/13/2023] [Accepted: 04/26/2024] [Indexed: 04/28/2024] Open
Abstract
The bone marrow is a specialized niche responsible for the maintenance of hematopoietic stem and progenitor cells during homeostasis and inflammation. Recent studies, however, have extended this essential role to the extramedullary and extravascular lung microenvironment. Here, we provide further evidence for a reservoir of hematopoietic stem and progenitor cells within the lung from Embryonic Day 18.5 until adulthood. These lung progenitors display distinct microenvironment-specific developmental kinetics compared with their bone marrow counterparts, exemplified by a rapid shift from a common myeloid to a megakaryocyte-erythrocyte progenitor-dominated niche with increasing age. In adult mice, influenza A viral infection results in a transient reduction in multipotent progenitors within the lungs, with a parallel increase in downstream granulocyte-monocyte progenitors and dendritic cell populations associated with acute viral infections. Our findings suggest that lung hematopoietic progenitors play a role in reestablishing immunological homeostasis in the respiratory mucosa, which may have significant clinical implications for maintaining pulmonary health after inflammatory perturbation.
Collapse
Affiliation(s)
- Kyle T Mincham
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jean-Francois Lauzon-Joset
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - James F Read
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona
| | - Patrick G Holt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia; and
| | - Philip A Stumbles
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, Western Australia, Australia
| | - Deborah H Strickland
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
8
|
Montante S, Ben-Othman R, Amenyogbe N, Angelidou A, van den Biggelaar A, Cai B, Chen Y, Darboe A, Diray-Arce J, Ford R, Idoko O, Lee A, Lo M, McEnaney K, Malek M, Martino D, Masiria G, Odumade OA, Pomat W, Shannon C, Smolen K, Consortium TEPIC, Ozonoff A, Richmond P, Tebbutt S, Levy O, Kampmann B, Brinkman R, Kollmann T. Breastfeeding and Neonatal Age Influence Neutrophil-Driven Ontogeny of Blood Cell Populations in the First Week of Human Life. J Immunol Res 2024; 2024:1117796. [PMID: 39081632 PMCID: PMC11288693 DOI: 10.1155/2024/1117796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/16/2024] [Accepted: 06/13/2024] [Indexed: 08/02/2024] Open
Abstract
The first few days of life are characterized by rapid external and internal changes that require substantial immune system adaptations. Despite growing evidence of the impact of this period on lifelong immune health, this period remains largely uncharted. To identify factors that may impact the trajectory of immune development, we conducted stringently standardized, high-throughput phenotyping of peripheral white blood cell (WBC) populations from 796 newborns across two distinct cohorts (The Gambia, West Africa; Papua New Guinea, Melanesia) in the framework of a Human Immunology Project Consortium (HIPC) study. Samples were collected twice from each newborn during the first week of life, first at Day of Life 0 (at birth) and then subsequently at Day of Life 1, 3, or 7 depending on the randomization group the newborn belongs to. The subsequent analysis was conducted at an unprecedented level of detail using flow cytometry and an unbiased automated gating algorithm. The results showed that WBC composition in peripheral blood changes along patterns highly conserved across populations and environments. Changes across days of life were most pronounced in the innate myeloid compartment. Breastfeeding, and at a smaller scale neonatal vaccination, were associated with changes in peripheral blood neutrophil and monocyte cell counts. Our results suggest a common trajectory of immune development in newborns and possible association with timing of breastfeeding initiation, which may contribute to immune-mediated protection from infection in early life. These data begin to outline a specific window of opportunity for interventions that could deliberately direct WBC composition, and with that, immune trajectory and thus ontogeny in early life. This trial is registered with NCT03246230.
Collapse
Affiliation(s)
| | - Rym Ben-Othman
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- RAN BioLinks Ltd., 10212 Yonge Street, 202, Richmond Hill L4C 3B6, Ontario, Canada
| | - Nelly Amenyogbe
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Department of Microbiology and ImmunologyDepartment of Pediatrics;Dalhousie University, 6299 South Street, Halifax B3H 4R2, Canada
| | - Asimenia Angelidou
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Department of NeonatologyBeth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Anita van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious DiseasesTelethon Kids InstituteUniversity of Western Australia Perth, 15 Hospital Avenue, Nedlands, WA 6009, Australia
| | - Bing Cai
- Department of PediatricsBC Children's HospitalUniversity of British Columbia, 4480 Oak Street, Vancouver V6H 3V4, BC, Canada
| | - Yixuan Chen
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
| | - Alansana Darboe
- Vaccines and Immunity ThemeMedical Research Council UnitThe Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul P.O. Box 273, Gambia
| | - Joann Diray-Arce
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Olubukola Idoko
- Department of Clinical ResearchFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Amy Lee
- Department of Molecular Biology and BiochemistrySimon Fraser University, 8888 University Dr. Burnaby V5A1S6, Burnaby, British Columbia, Canada
| | - Mandy Lo
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
| | - Kerry McEnaney
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
| | - Mehrnoush Malek
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
| | - David Martino
- Wal-yan Respiratory Research CentreTelethon Kids InstituteUniversity of Western Australia, Perth, Australia
| | - Geraldine Masiria
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Oludare A. Odumade
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - William Pomat
- Papua New Guinea Institute of Medical Research, Homate Street, 441, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Casey Shannon
- PROOF Centre of Excellence, 10th floor, 1190 Hornby Street, Vancouver V6Z 2K5, British Columbia, Canada
- UBC Centre for Heart Lung InnovationSt. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Kinga Smolen
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Al Ozonoff
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter Richmond
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Division of PediatricsSchool of MedicineUniversity of Western Australia, 35 Stirling Highway, Crawley 6009, WA, Australia
| | - Scott Tebbutt
- PROOF Centre of Excellence, 10th floor, 1190 Hornby Street, Vancouver V6Z 2K5, British Columbia, Canada
- UBC Centre for Heart Lung InnovationSt. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Ofer Levy
- Precision Vaccines ProgramDepartment of PediatricsBoston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beate Kampmann
- Vaccines and Immunity ThemeMedical Research Council UnitThe Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul P.O. Box 273, Gambia
- Centre for Global Health and Institute for International HealthCharite Universitatsmedizin, Berlin, Germany
| | - Ryan Brinkman
- BC Cancer Agency, 675 West 10th Avenue, Vancouver V5Z 1G1, BC, Canada
- Department of Medical GeneticsUniversity of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia V6T1Z4, Canada
| | - Tobias Kollmann
- Telethon Kids InstitutePerth Children's Hospital, 15 Hospital Avenue, Nedlands 6009, WA, Australia
- Department of PediatricsBC Children's HospitalUniversity of British Columbia, 4480 Oak Street, Vancouver V6H 3V4, BC, Canada
- Microbiology and ImmunologyPediatric Infectious DiseasesDalhousie University, CEO, Born, Strong Initiative, Halifax, Nova Scotia, Canada
| |
Collapse
|
9
|
Wolska M, Wypych TP, Rodríguez-Viso P. The Influence of Premature Birth on the Development of Pulmonary Diseases: Focus on the Microbiome. Metabolites 2024; 14:382. [PMID: 39057705 PMCID: PMC11279213 DOI: 10.3390/metabo14070382] [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: 06/06/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Globally, around 11% of neonates are born prematurely, comprising a highly vulnerable population with a myriad of health problems. Premature births are often accompanied by an underdeveloped immune system biased towards a Th2 phenotype and microbiota dysbiosis. Typically, a healthy gut microbiota interacts with the host, driving the proper maturation of the host immunity. However, factors like cesarean section, formula milk feeding, hospitalization in neonatal intensive care units (NICU), and routine antibiotic treatments compromise microbial colonization and increase the risk of developing related diseases. This, along with alterations in the innate immune system, could predispose the neonates to the development of respiratory diseases later in life. Currently, therapeutic strategies are mainly focused on restoring gut microbiota composition using probiotics and prebiotics. Understanding the interactions between the gut microbiota and the immature immune system in premature neonates could help to develop novel therapeutic strategies for treating or preventing gut-lung axis disorders.
Collapse
Affiliation(s)
| | - Tomasz Piotr Wypych
- Laboratory of Host-Microbiota Interactions, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Ludwika Pasteura 3, 02-093 Warsaw, Poland; (M.W.); (P.R.-V.)
| | | |
Collapse
|
10
|
Chen Y, Lu C, Huang J, Li L, Yang Y, Shao Y, Liu L, Sun B. Characteristics of Neonatal Sepsis and Predictive Values of Polyfunctional Assessment of Umbilical Cord Neutrophils Based on Single Cell Proteomic Secretion. Inflammation 2024:10.1007/s10753-024-02095-2. [PMID: 38976117 DOI: 10.1007/s10753-024-02095-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/04/2024] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
The early diagnosis of neonatal sepsis is crucial as it remains a prevalent cause of neonatal mortality. In this study, we conducted an analysis on the clinical data and detection indicators of 22 cases with sepsis and 62 cases without sepsis among neonates. Our findings indicate that the clinical signs observed in neonates with sepsis lack specificity. In addition, the commonly used clinical inflammatory indicators (such as leukocyte count, neutrophil-to-lymphocyte ratio [NLR], C-reactive protein [CRP], procalcitonin) exhibit limited sensitivity and specificity. Furthermore, the current clinical measures lack the assessment of inflammatory factors. Therefore, in order to enhance the accuracy of early sepsis diagnosis in neonates, we have employed a novel microfluidic-based single-cell technology platform for the analysis of 32 cytokines secreted by neutrophils at the individual cell level under various toxin stimulation conditions. We have further investigated and compared the disparities in single-cell protein secretomics between umbilical cord blood neutrophils and healthy adult peripheral neutrophils within an in vitro sepsis model. Our findings indicate that in a resting state UCB neutrophils exhibited lower polyfunctionality compared with healthy adult blood neutrophils, and notable variations in cytokine secretion profiles were detected between the two groups. However, the polyfunctionality of UCB neutrophils significantly increased and surpassed that of healthy adult neutrophils when exposed to alpha-hemolysin or lipopolysaccharide. UCB neutrophils secreted a wide range of chemokines and inflammatory factors, among which GM-CSF and IL-18 were the most significant. Furthermore, we initially categorized the functional subgroups of neutrophils by considering the secretion of five primary cytokines by neutrophils (GM-CSF, IL-18, IL-8, MIP-1β, and MIF). The current study, for the first time, examined in detail the heterogeneity of protein secretion and the functional diversity of UCB neutrophils stimulated by different antigens. Moreover, new insight into neonatal sepsis, early diagnosis, and wider clinical applications of UCB neutrophils are provided by these data.
Collapse
Affiliation(s)
- Yi Chen
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Burns and Plastic Surgery, Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Cheng Lu
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jiamin Huang
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Linbin Li
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yunxi Yang
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lu Liu
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Bingwei Sun
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
11
|
López-Arredondo A, Cruz-Cardenas JA, Cázares-Preciado JA, Timmins NE, Brunck ME. Neutrophils, an emerging new therapeutic platform. Curr Opin Biotechnol 2024; 87:103106. [PMID: 38490109 DOI: 10.1016/j.copbio.2024.103106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
Neutrophils possess unique characteristics that render them indispensable to health, and patients with irregular neutrophil counts or functions suffer from increased morbidity and mortality. As neutrophils are short-lived postmitotic cells, genetic aberrations cannot be corrected directly in neutrophils and must be targeted in their progenitors. Neutrophils are increasingly being contemplated for a range of therapeutic applications, including restoration or modulation of immune function and targeting of solid tumors. This review addresses the state-of-the-art in neutrophil transfusions and their possible applications for infectious disease prevention and treatment. It offers a landscape of the most recent gene therapy approaches to address neutrophil-related genetic diseases. We also discuss how ongoing research could broaden the applicability of neutrophil-based therapies to solid cancer treatments and beyond.
Collapse
Affiliation(s)
- Alejandra López-Arredondo
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849 Monterrey, Nuevo León, Mexico
| | - José A Cruz-Cardenas
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849 Monterrey, Nuevo León, Mexico
| | - Jorge A Cázares-Preciado
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849 Monterrey, Nuevo León, Mexico
| | - Nicholas E Timmins
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Marion Eg Brunck
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849 Monterrey, Nuevo León, Mexico; The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnologico, 64849 Monterrey, Nuevo León, Mexico.
| |
Collapse
|
12
|
Joyce LR, Kim S, Spencer BL, Christensen PM, Palmer KL, Guan Z, Siegenthaler JA, Doran KS. Streptococcus agalactiae glycolipids promote virulence by thwarting immune cell clearance. SCIENCE ADVANCES 2024; 10:eadn7848. [PMID: 38809989 PMCID: PMC11135403 DOI: 10.1126/sciadv.adn7848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/25/2024] [Indexed: 05/31/2024]
Abstract
Streptococcus agalactiae [group B Streptococcus (GBS)] is a leading cause of neonatal meningitis, with late-onset disease (LOD) occurring after gastrointestinal tract colonization in infants. Bacterial membrane lipids are essential for host-pathogen interactions, and the functions of glycolipids are yet to be fully elucidated. GBS synthesizes three major glycolipids: glucosyl-diacylglycerol (Glc-DAG), diglucosyl-DAG (Glc2-DAG), and lysyl-Glc-DAG (Lys-Glc-DAG). Here, we identify the enzyme, IagB, as responsible for biosynthesis of Glc-DAG, the precursor for the two other glycolipids in GBS. To examine the collective role of glycolipids to GBS virulence, we adapted a murine model of neonatal meningitis to simulate LOD. The GBS∆iagB mutant traversed the gut-epithelial barrier comparable to wild type but was severely attenuated in bloodstream survival, resulting in decreased bacterial loads in the brain. The GBS∆iagB mutant was more susceptible to neutrophil killing and membrane targeting by host antimicrobial peptides. This work reveals an unexplored function of GBS glycolipids with their ability to protect the bacterial cell from host antimicrobial killing.
Collapse
Affiliation(s)
- Luke R. Joyce
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sol Kim
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Priya M. Christensen
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Kelli L. Palmer
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Julie A. Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
13
|
Grudzinska FS, Scott A. Unravelling the neutrophil enigma: a new insight into RSV-induced bronchiolitis in infants. Thorax 2024; 79:487-488. [PMID: 38575318 DOI: 10.1136/thorax-2023-221130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 04/06/2024]
Affiliation(s)
- Frances Susanna Grudzinska
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Aaron Scott
- Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
14
|
Rooke Z, Zainal Abidin N, Harris C, Brodlie M. Neutrophils and RSV: differences with age. Thorax 2024; 79:489-490. [PMID: 38388487 DOI: 10.1136/thorax-2023-221177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Affiliation(s)
- Zoe Rooke
- Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Noreen Zainal Abidin
- Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Caroline Harris
- Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Malcolm Brodlie
- Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Parsons EL, Kim JS, Malloy AMW. Development of innate and adaptive immunity to RSV in young children. Cell Immunol 2024; 399-400:104824. [PMID: 38615612 DOI: 10.1016/j.cellimm.2024.104824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Infection of the respiratory tract with respiratory syncytial virus (RSV) is common and occurs repeatedly throughout life with most severe disease occurring at the extremes of age: in young infants and the elderly. Effective anti-viral therapeutics are not available and therefore prevention has been the primary strategy for reducing the disease burden. Our current understanding of respiratory mucosal cell biology and the immune response within the respiratory tract is inadequate to prevent infection caused by a pathogen like RSV that does not disseminate outside of this environment. Gaps in our understanding of the activation of innate and adaptive immunity in response to RSV and the role of age upon infection also limit improvements in the design of therapeutics and vaccines for young infants. However, advancements in structural biology have improved our ability to characterize antibodies against viral proteins and in 2023 the first vaccines for those over 60 years and pregnant women became available, potentially reducing the burden of disease. This review will examine our current understanding of the critical facets of anti-RSV immune responses in infants and young children as well as highlight areas where more research is needed.
Collapse
Affiliation(s)
| | - Jisung S Kim
- Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation, Bethesda, MD, USA
| | | |
Collapse
|
16
|
Miková E, Černý V, Novotná O, Petrásková P, Boráková K, Hel Z, Hrdý J. Immature neutrophils in cord blood exert increased expression of genes associated with antimicrobial function. Front Immunol 2024; 15:1368624. [PMID: 38596677 PMCID: PMC11002259 DOI: 10.3389/fimmu.2024.1368624] [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: 01/10/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction The immune systems of both the mother and the newborn face significant challenges during birth. Proper immune regulation after birth is essential for the survival of neonates. Numerous studies have demonstrated that the neonatal immune system is relatively immature, particularly in its adaptive arm, placing the primary responsibility for immune surveillance on innate immunity. Methods Given the significant role of neutrophils in protecting the neonate after birth, we conducted a study investigating the properties of neutrophils in newborn cord blood using various methodological approaches. Results Our findings demonstrate the presence of immature low-density neutrophils in the cord blood, which are likely responsible for the observed elevated expression of genes coding for proteins essential to antimicrobial response, including myeloperoxidase, neutrophils elastase, and defensins. Discussion We propose that these cells function normally and support the protection of newborns early after birth. Furthermore, our results suggest that the mode of delivery might significantly influence the programming of neutrophil function. The presented findings emphasize the importance of distinct neutrophil subpopulations in neonatal immunity and their potential impact on early postnatal health.
Collapse
Affiliation(s)
- Eliška Miková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Viktor Černý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Olga Novotná
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Petra Petrásková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Kristýna Boráková
- Department of Neonatology, Institute for the Care of Mother and Child, Prague, Czechia
| | - Zdenek Hel
- Pathology Department, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
17
|
Collins A, Swann JW, Proven MA, Patel CM, Mitchell CA, Kasbekar M, Dellorusso PV, Passegué E. Maternal inflammation regulates fetal emergency myelopoiesis. Cell 2024; 187:1402-1421.e21. [PMID: 38428422 PMCID: PMC10954379 DOI: 10.1016/j.cell.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/03/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
Neonates are highly susceptible to inflammation and infection. Here, we investigate how late fetal liver (FL) mouse hematopoietic stem and progenitor cells (HSPCs) respond to inflammation, testing the hypothesis that deficits in the engagement of emergency myelopoiesis (EM) pathways limit neutrophil output and contribute to perinatal neutropenia. We show that fetal HSPCs have limited production of myeloid cells at steady state and fail to activate a classical adult-like EM transcriptional program. Moreover, we find that fetal HSPCs can respond to EM-inducing inflammatory stimuli in vitro but are restricted by maternal anti-inflammatory factors, primarily interleukin-10 (IL-10), from activating EM pathways in utero. Accordingly, we demonstrate that the loss of maternal IL-10 restores EM activation in fetal HSPCs but at the cost of fetal demise. These results reveal the evolutionary trade-off inherent in maternal anti-inflammatory responses that maintain pregnancy but render the fetus unresponsive to EM activation signals and susceptible to infection.
Collapse
Affiliation(s)
- Amélie Collins
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Neonatology-Perinatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - James W Swann
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Melissa A Proven
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chandani M Patel
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Carl A Mitchell
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Monica Kasbekar
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Hematology/Oncology, Department of Internal Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
18
|
Rühle J, Schwarz J, Dietz S, Rückle X, Schoppmeier U, Lajqi T, Poets CF, Gille C, Köstlin-Gille N. Impact of perinatal administration of probiotics on immune cell composition in neonatal mice. Pediatr Res 2024:10.1038/s41390-024-03029-2. [PMID: 38278847 DOI: 10.1038/s41390-024-03029-2] [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] [Received: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Newborns and especially preterm infants are much more susceptible to infections than adults. The pathogens causing infections in newborns are often detectable in the intestinal flora of affected children even before disease onset. Therefore, it seems reasonable to prevent dysbiosis in newborns and preterm infants. An approach followed in many neonatal intensive care units (NICUs) is to prevent infections in preterm infants with probiotics however their mechanisms of action of probiotics are incompletely understood. Here, we investigated the effect of perinatal probiotic exposure on immune cells in newborn mice. METHODS Pregnant mice were orally treated with a combination of Lactobacillus acidophilus and Bifidobacterium bifidum (Infloran®) from mid-pregnancy until the offspring were harvested. Immune cell composition in organs of the offspring were analyzed by flow cytometry. RESULTS Perinatal probiotic exposure had profound effects on immune cell composition in the intestine, liver and lungs of newborn mice with reduction of myeloid and B cells and induction of T cells in the probiotic treated animals' organs at weaning. Furthermore, probiotic exposure had an effect on T cell development in the thymus. CONCLUSION Our results contribute to a better understanding of the interaction of probiotics with the developing immune system. IMPACT probiotics have profound effects on immune cell composition in intestines, livers and lungs of newborn mice. probiotics modulate T cell development in thymus of newborn mice. effects of probiotics on neonatal immune cells are particularly relevant in transition phases of the microbiome. our results contribute to a better understanding of the mechanisms of action of probiotics in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Xenia Rückle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Ulrich Schoppmeier
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| |
Collapse
|
19
|
Vergadi E, Kolliniati O, Lapi I, Ieronymaki E, Lyroni K, Alexaki VI, Diamantaki E, Vaporidi K, Hatzidaki E, Papadaki HA, Galanakis E, Hajishengallis G, Chavakis T, Tsatsanis C. An IL-10/DEL-1 axis supports granulopoiesis and survival from sepsis in early life. Nat Commun 2024; 15:680. [PMID: 38263289 PMCID: PMC10805706 DOI: 10.1038/s41467-023-44178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/03/2023] [Indexed: 01/25/2024] Open
Abstract
The limited reserves of neutrophils are implicated in the susceptibility to infection in neonates, however the regulation of neutrophil kinetics in infections in early life remains poorly understood. Here we show that the developmental endothelial locus (DEL-1) is elevated in neonates and is critical for survival from neonatal polymicrobial sepsis, by supporting emergency granulopoiesis. Septic DEL-1 deficient neonate mice display low numbers of myeloid-biased multipotent and granulocyte-macrophage progenitors in the bone marrow, resulting in neutropenia, exaggerated bacteremia, and increased mortality; defects that are rescued by DEL-1 administration. A high IL-10/IL-17A ratio, observed in newborn sepsis, sustains tissue DEL-1 expression, as IL-10 upregulates while IL-17 downregulates DEL-1. Consistently, serum DEL-1 and blood neutrophils are elevated in septic adult and neonate patients with high serum IL-10/IL-17A ratio, and mortality is lower in septic patients with high serum DEL-1. Therefore, IL-10/DEL-1 axis supports emergency granulopoiesis, prevents neutropenia and promotes sepsis survival in early life.
Collapse
Affiliation(s)
- Eleni Vergadi
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece.
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece.
| | - Ourania Kolliniati
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Ioanna Lapi
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Ieronymaki
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Konstantina Lyroni
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Eleni Diamantaki
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Eleftheria Hatzidaki
- Department of Neonatology/Neonatal Intensive Care Unit, School of Medicine, University of Crete, Heraklion, Greece
| | - Helen A Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece
| | - Emmanouil Galanakis
- Department of Paediatrics, School of Medicine, University of Crete, Heraklion, Greece
| | - George Hajishengallis
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christos Tsatsanis
- Institute of Molecular Biology and Biotechnology, IMMB, FORTH, Heraklion, Greece
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
20
|
Li Z, Yuan T. Neutrophil extracellular traps in adult diseases and neonatal bacterial infectious diseases: A review. Heliyon 2024; 10:e23559. [PMID: 38173520 PMCID: PMC10761809 DOI: 10.1016/j.heliyon.2023.e23559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Neutrophils, the most abundant type of white blood cells, are pivotal in fighting bacterial infections due to their immunological and anti-infection capabilities. In recent years, scientists have discovered a novel mechanism known as neutrophil extracellular traps, which are fibrous networks primarily released by neutrophils that combat bacterial infections. There is a growing interest in studying NETs and their role in human infectious diseases, particularly in neonates susceptible to bacterial infections. NETs and their components have been found in various samples from neonatal-infected patients, providing a new route for early diagnosis of neonatal infectious diseases. This paper aims to summarize the studies on NETs in adult diseases and mainly discuss NETs in neonatal sepsis, necrotizing enterocolitis, and purulent meningitis, to provide scientific evidence for early monitoring, diagnosis, and treatment of neonatal infections.
Collapse
Affiliation(s)
- Ziheng Li
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| | - Tianming Yuan
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, China
| |
Collapse
|
21
|
Ledderose C, Valsami EA, Newhams M, Elevado MJ, Novak T, Randolph AG, Junger WG. ATP breakdown in plasma of children limits the antimicrobial effectiveness of their neutrophils. Purinergic Signal 2023; 19:651-662. [PMID: 36596963 PMCID: PMC10754799 DOI: 10.1007/s11302-022-09915-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023] Open
Abstract
Neutrophils (PMNs) require extracellular ATP and adenosine (ADO) to fight bacterial infections, which often have life-threatening consequences in pediatric patients. We wondered whether the ATP and ADO levels in the plasma of children change with age and if these changes influence the antimicrobial efficacy of the PMNs of these children. We measured plasma concentrations of ATP and ADO and the activities of the enzymes responsible for the breakdown of these mediators in plasma samples from healthy children and adolescents (n = 45) ranging in age from 0.2 to 15 years. In addition, using blood samples of these individuals, we compared how effective their PMNs were in the phagocytosis of bacteria. In an experimental sepsis model with young (10 days) and adolescent mice (10 weeks), we studied how age influenced the resilience of these animals to bacterial infections and whether addition of ATP could improve the antimicrobial capacity of their PMNs. We found that plasma ATP levels correlated with age and were significantly lower in infants (< 1 year) than in adolescents (12-15 years). In addition, we observed significantly higher plasma ATPase and adenosine deaminase activities in children (< 12 years) when compared to the adolescent population. The activities of these ATP and ADO breakdown processes correlated inversely with age and with the ability of PMNs to phagocytize bacteria. Similar to their human counterparts, young mice also had significantly lower plasma ATP levels when compared to adolescent animals. In addition, we found that mortality of young mice after bacterial infection was significantly higher than that of adolescent mice. Moreover, bacterial phagocytosis by PMNs of young mice was weaker when compared to that of older mice. Finally, we found that ATP supplementation could recover bacterial phagocytosis of young mice to levels similar to those of adolescent mice. Our findings suggest that rapid ATP hydrolysis in the plasma of young children lowers the antimicrobial functions of their PMNs and that this may contribute to the vulnerability of pediatric patients to bacterial infections.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Eleftheria-Angeliki Valsami
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Margaret Newhams
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark-Josef Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
22
|
Şişmanlar Eyuboglu T, Aslan AT, Asfuroglu P, Kunt N, Ersoy A, Kose M, Unal G, Pekcan S. Neutrophil lymphocyte ratio, mean platelet volume, and immunoreactive trypsinogen as early inflammatory biomarkers for cystic fibrosis in infancy: A retrospective cohort study. Pediatr Pulmonol 2023; 58:3106-3112. [PMID: 37530491 DOI: 10.1002/ppul.26628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/27/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Airway inflammation starts in early life in cystic fibrosis (CF) and limited, objective markers are available to help identify infants with increased inflammation. We aimed to investigate neutrophil, lymphocyte ratio (NLR), mean platelet volume (MPV) and immunoreactive trypsinogen (IRT) to be a possible inflammatory biomarker for CF in infancy. METHODS This was a retrospective cohort study in three centers. Between January 2015 and December 2022, children with CF newborn screening (NBS) positivity and diagnosed as CF were included in the study. Correlation analysis were performed with NLR, MPV, IRT and follow-up parameters such as z-scores, modified Shwachman-Kulczycki score (mSKS) at the first, second, third and sixth ages and pulmonary function test (PFT) at the sixth age. RESULTS A total of 92 children with CF included in the study and 47.8% of them were female. There were no correlations between NLR, MPV and weight and height z-scores for all ages (p > 0.05), a negative correlation was found between MPV and body mass indexes (BMI) z-score at the age of 6 (r = -0.443, p = 0.038). No correlation was found between NLR, MPV and PFT parameters and mSKS at all ages (p > 0.05). There was a negative correlation between first IRT and BMI z-score at 6 years of age (r = -0.381, p = 0.046) and negative correlations between second IRT and weight and BMI z-score at the age of 6 (r = -0.462, p = 0.010; r = -0.437, p = 0.016, respectively). CONCLUSION Higher MPV and IRT levels during NBS period are associated with worse nutritional outcome which may reflect chronic inflammation. Children with higher MPV and IRT should be followed up closely in terms of chronic inflammation and nutritional status.
Collapse
Affiliation(s)
| | - Ayse Tana Aslan
- Department of Pediatric Pulmonology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Pelin Asfuroglu
- Department of Pediatric Pulmonology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Nursima Kunt
- Department of Pediatrics, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ali Ersoy
- Department of Pediatric Pulmonology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Mehmet Kose
- Department of Pediatric Pulmonology, Erciyes University Faculty of Medicine, Kayseri, Turkey
| | - Gokcen Unal
- Department of Pediatric Pulmonology, Necmettin Erbakan University Meram Medicine Faculty, Konya, Turkey
| | - Sevgi Pekcan
- Department of Pediatric Pulmonology, Necmettin Erbakan University Meram Medicine Faculty, Konya, Turkey
| |
Collapse
|
23
|
Liang X, Luo H, Chen X, Yu X, Yan Q. Prognostic value of blood inflammatory parameters for predicting mortality in neonates with sepsis. Int Immunopharmacol 2023; 123:110780. [PMID: 37572503 DOI: 10.1016/j.intimp.2023.110780] [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: 04/27/2023] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Neonatal sepsis is a major cause of morbidity and mortality in neonates. The diagnosis of neonatal sepsis has been widely explored using blood inflammatory parameters. However, few researches have focused on the predictive significance of blood inflammation parameters for predicting mortality. This study aimed to evaluate the prognostic value of blood inflammatory parameters, including white blood cell (WBC), neutrophil, lymphocyte, monocyte, platelet and C-reactive protein (CRP) for predicting mortality in neonates with sepsis. METHODS Neonates with culture-proven sepsis were enrolled in this study. The clinical characteristics and levels of white blood cell, neutrophil, lymphocyte, monocyte, platelet and CRP were recorded. The receiver-operating characteristic (ROC) curve was applied to calculate the area under the curve (AUC) and determine the optimal cutoff values. Multivariable Cox regression model was used to evaluate the independent prognostic significance of variables. Kaplan-Meier curve was used to assess survival. RESULTS A total of 188 neonates with culture-proven sepsis were included for analysis. The 7-day mortality rate was 11.2 % (21/188) and the 28-day mortality rate was 13.8 % (26/188). The levels of white blood cell, neutrophil, monocyte and platelet in non-survivors were lower than those in survivors (P < 0.05). Platelet yielded higher AUC values than other parameters for predicting mortality with the best cutoff value of 132 × 109/L, followed by WBC with the optimal cutoff value of 6.15 × 109/L. Multivariable Cox regression analysis showed platelet and WBC were independent prognostic factors for predicting mortality. Low platelet group showed lower survival according to Kaplan-Meier method. CONCLUSIONS In conclusion, the levels of platelet and WBC on the day of sepsis onset are valuable indicators for predicting mortality in neonates with sepsis.
Collapse
Affiliation(s)
- Xianghui Liang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huijuan Luo
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xia Chen
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohe Yu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Grabbe M, Conejeros I, Velásquez ZD, Hasheminasab SS, Kamena F, Wehrend A, Gärtner U, Taubert A, Hermosilla CR. Cryptosporidium parvum-induced neutrophil extracellular traps in neonatal calves is a stage-independent process. Front Vet Sci 2023; 10:1256726. [PMID: 37662980 PMCID: PMC10470472 DOI: 10.3389/fvets.2023.1256726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction Infections with the apicomplexan obligate intracellular parasite Cryptosporidium parvum lead to cryptosporidiosis-a worldwide zoonotic infection. C. parvum is one of the most common diarrheal pathogens in young calves, which are the main reservoir of the pathogen. Cryptosporidiosis leads to severe economic losses in the calf industry and being a major contributor to diarrhea morbidity and mortality in children. Polymorphonuclear neutrophils (PMN) are part of the innate immune system. Their effector mechanisms directed against invasive parasites include phagocytosis, production of antimicrobial molecules as well as the formation of so-called neutrophil extracellular traps (NETs). Like other leukocytes of the innate immune system, PMN are thus able to release chromatin fibers enriched with antimicrobial granular molecules extracellularly thereby immobilizing and partially killing invasive bacteria, viruses, fungi and parasites. Methods In vitro interactions of neonatal bovine PMN and C. parvum-oocysts and sporozoites were illustrated microscopically via scanning electron microscopy- and live cell imaging 3D holotomographic microscopy analyses. C. parvum-triggered NETosis was quantified via extracellular DNA measurements as well as verified via detection of NET-typical molecules [histones, neutrophil elastase (NE)] through immunofluorescence microscopy analysis. To verify the role of ATP in neonatal-derived NETosis, inhibition experiments were performed with NF449 (purinergic receptor antagonist with high specificity to P2X1 receptor). Results and discussion Using immunofluorescence- and SEM-based analyses, we demonstrate here for the first time that neonate bovine PMN are capable of forming NETs against C. parvum-sporozoites and oocysts, thus as a stage-independent cell death process. Our data further showed that C. parvum strongly induces suicidal neonatal NETosis in a P2X1-dependent manner, suggesting anti-cryptosporidial effects not only through firm sporozoite ensnarement and hampered sporozoite excystation, but also via direct exposure to NETs-associated toxic components.
Collapse
Affiliation(s)
- Magdalena Grabbe
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| | - Iván Conejeros
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| | - Zahady D. Velásquez
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| | - Seyed Sajjad Hasheminasab
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| | - Faustin Kamena
- Laboratory for Molecular Parasitology, Department of Microbiology and Parasitology, University of Buea, Buea, Cameroon
| | - Axel Wehrend
- Clinic for Obstetrics, Gynaecology and Andrology of Large and Small Animals With Veterinary Ambulance, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Rodrigo Hermosilla
- Institute of Parasitology, Biomedical Center Seltersberg (BFS), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
25
|
Rühle J, Ginzel M, Dietz S, Schwarz J, Lajqi T, Beer-Hammer S, Poets CF, Gille C, Köstlin-Gille N. Depletion of Ly6G-Expressing Neutrophilic Cells Leads to Altered Peripheral T-Cell Homeostasis and Thymic Development in Neonatal Mice. Int J Mol Sci 2023; 24:7763. [PMID: 37175470 PMCID: PMC10178674 DOI: 10.3390/ijms24097763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Newborns and especially preterm infants are much more susceptible to infections than adults. Due to immature adaptive immunity, especially innate immune cells play an important role in a newborn's infection defense. Neonatal neutrophils exhibit profound differences in their functionality compared to neutrophils of adults. In particular, neonates possess a relevant population of suppressive neutrophils, which not only inhibit but also specifically modulate the function of T-cells. In this study, we investigated whether neonatal neutrophils are already involved in T-cell development in the thymus. For this purpose, we used a newly developed model of antibody-mediated immune cell depletion in which we administered a depleting antibody to pregnant and then lactating dams. Using this method, we were able to sufficiently deplete Ly6G-positive neutrophils in offspring. We demonstrated that the depletion of neutrophils in newborn mice resulted in altered peripheral T-cell homeostasis with a decreased CD4+/CD8+ T-cell ratio and decreased expression of CD62L. Neutrophil depletion even affected T-cell development in the thymus, with increased double positive thymocytes and a decreased CD4+/CD8+ single positive thymocyte ratio. Altogether, we demonstrated a previously unknown mechanism mediating neutrophils' immunomodulatory effects in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Marco Ginzel
- Department of Pediatric and Adolescent Surgery, Paracelsus Medical University Hospital, A-5020 Salzburg, Austria;
| | - Stefanie Dietz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Julian Schwarz
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomic and ICePhA, University of Tübingen, D-72074 Tübingen, Germany;
| | - Christian F. Poets
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| | - Natascha Köstlin-Gille
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany; (J.R.); (S.D.); (J.S.); (C.F.P.)
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany; (T.L.); (C.G.)
| |
Collapse
|
26
|
Neonatal hematological parameters: the translational aspect of developmental hematopoiesis. Ann Hematol 2023; 102:707-714. [PMID: 36847806 DOI: 10.1007/s00277-023-05144-2] [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: 06/17/2022] [Accepted: 12/20/2022] [Indexed: 03/01/2023]
Abstract
Hematopoiesis is a process constantly evolving from fetal life through adulthood. Neonates present with qualitative and quantitative differences in hematological parameters compared to older children and adults, reflecting developmental changes in hematopoiesis correlated with gestational age. Such differences are more intense for preterm and small-for-gestational-age neonates or neonates with intrauterine growth restriction. This review article is aimed at describing the hematologic differences among neonatal subgroups and the major underlying pathogenic mechanisms. Issues that should be taken into account when interpreting neonatal hematological parameters are also highlighted.
Collapse
|
27
|
Bernis ME, Zweyer M, Maes E, Schleehuber Y, Sabir H. Neutrophil Extracellular Traps Release following Hypoxic-Ischemic Brain Injury in Newborn Rats Treated with Therapeutic Hypothermia. Int J Mol Sci 2023; 24:3598. [PMID: 36835009 PMCID: PMC9966013 DOI: 10.3390/ijms24043598] [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: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
The peripheral immune system plays a critical role in neuroinflammation of the central nervous system after an insult. Hypoxic-ischemic encephalopathy (HIE) induces a strong neuroinflammatory response in neonates, which is often associated with exacerbated outcomes. In adult models of ischemic stroke, neutrophils infiltrate injured brain tissue immediately after an ischemic insult and aggravate inflammation via various mechanisms, including neutrophil extracellular trap (NETs) formation. In this study, we used a neonatal model of experimental hypoxic-ischemic (HI) brain injury and demonstrated that circulating neutrophils were rapidly activated in neonatal blood. We observed an increased infiltration of neutrophils in the brain after exposure to HI. After treatment with either normothermia (NT) or therapeutic hypothermia (TH), we observed a significantly enhanced expression level of the NETosis marker Citrullinated H3 (Cit-H3), which was significantly more pronounced in animals treated with TH than in those treated with NT. NETs and NLR family pyrin domain containing 3 (NLRP-3) inflammasome assembly are closely linked in adult models of ischemic brain injury. In this study, we observed an increase in the activation of the NLRP-3 inflammasome at the time points analyzed, particularly immediately after TH, when we observed a significant increase in NETs structures in the brain. Together, these results suggest the important pathological functions of early arriving neutrophils and NETosis following neonatal HI, particularly after TH treatment, which is a promising starting point for the development of potential new therapeutic targets for neonatal HIE.
Collapse
Affiliation(s)
- Maria E. Bernis
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Elke Maes
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Yvonne Schleehuber
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, 53127 Bonn, Germany
- Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| |
Collapse
|
28
|
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
|
29
|
Okazaki K, Nakamura S, Koyano K, Konishi Y, Kondo M, Kusaka T. Neonatal asphyxia as an inflammatory disease: Reactive oxygen species and cytokines. Front Pediatr 2023; 11:1070743. [PMID: 36776908 PMCID: PMC9911547 DOI: 10.3389/fped.2023.1070743] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Neonatologists resuscitate asphyxiated neonates by every available means, including positive ventilation, oxygen therapy, and drugs. Asphyxiated neonates sometimes present symptoms that mimic those of inflammation, such as fever and edema. The main pathophysiology of the asphyxia is inflammation caused by hypoxic-ischemic reperfusion. At birth or in the perinatal period, neonates may suffer several, hypoxic insults, which can activate inflammatory cells and inflammatory mediator production leading to the release of larger quantities of reactive oxygen species (ROS). This in turn triggers the production of oxygen stress-induced high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular patterns (DAMPs) protein bound to toll-like receptor (TLR) -4, which activates nuclear factor-kappa B (NF-κB), resulting in the production of excess inflammatory mediators. ROS and inflammatory mediators are produced not only in activated inflammatory cells but also in non-immune cells, such as endothelial cells. Hypothermia inhibits pro-inflammatory mediators. A combination therapy of hypothermia and medications, such as erythropoietin and melatonin, is attracting attention now. These medications have both anti-oxidant and anti-inflammatory effects. As the inflammatory response and oxidative stress play a critical role in the pathophysiology of neonatal asphyxia, these drugs may contribute to improving patient outcomes.
Collapse
Affiliation(s)
- Kaoru Okazaki
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kosuke Koyano
- Maternal Perinatal Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Masatoshi Kondo
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
30
|
Miller NW, Seman BG, Akers SM, Povroznik JM, Brundage K, Fang W, Robinson CM. The impact of opioid exposure during pregnancy on the human neonatal immune profile. Pediatr Res 2022; 92:1566-1574. [PMID: 35288639 PMCID: PMC8920062 DOI: 10.1038/s41390-022-02014-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/19/2022] [Accepted: 02/04/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The increasing magnitude of the opioid crisis and rising rates of neonatal abstinence syndrome (NAS) diagnoses highlight the need for increased research into how maternal substance use during pregnancy can impact the neonatal immune profile and its functionality. We hypothesized that neonates with opioid exposure would have reduced proportions of some immune cells, an anti-inflammatory cytokine profile, reduced T cell proliferation, and monocyte bacterial killing activity compared to the control population. METHODS The present study compares immune cell populations, inflammatory and anti-inflammatory cytokine and chemokine levels in the serum, and monocyte and T cell functional activity using umbilical cord samples from neonates with known opioid exposure during gestation and from control neonates without known exposure. RESULTS Our findings demonstrated a significant reduction in neutrophils, decreased levels of inflammatory cytokines in the serum, and reduced IL-2 production during in vitro CD4+ T cell proliferation in neonates exposed to opioids compared to controls. The neutrophil findings were supported by retrospective analysis of an extended network of deidentified patient records. CONCLUSIONS This study is the first of its kind to evaluate differences in neonatal immunity as a result of opioid exposure in the human population that will inform continued mechanistic studies. IMPACT The opioid epidemic has become a public health crisis in the United States, and the corresponding incidence of neonatal abstinence syndrome (NAS) have risen accordingly. New research is required to understand the short and long-term health impacts of opioid exposure to the neonate. This is the first human study to investigate the immunologic profile and functionality in neonates with known opioid exposure in utero. The abundance of neutrophils and the ratio of neutrophils to lymphocytes is significantly reduced along with inflammatory cytokines and chemokines following opioid exposure during pregnancy. The immune profile in opioid-exposed neonates may promote susceptibility to infection.
Collapse
Affiliation(s)
- Nicholas W Miller
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Brittany G Seman
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Stephen M Akers
- Department of Pediatrics, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jessica M Povroznik
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA
| | - Kathleen Brundage
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Wei Fang
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, 26506, USA
| | - Cory M Robinson
- Department of Microbiology, Immunology, & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| |
Collapse
|
31
|
Unal S, Demirel N, Arslan Z, Tokgoz-Cuni B, Ulubas-Isik D, Bas AY. Umbilical Cord Separation Time and Influencing Factors in Very-Low-Birth-Weight Preterm Neonates. Am J Perinatol 2022; 39:1682-1687. [PMID: 33657638 DOI: 10.1055/s-0041-1726035] [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: 10/22/2022]
Abstract
OBJECTIVE The average time for umbilical cord separation in term neonates is 7 to 10 days. Prematurity, phototherapy, prolonged duration of antibiotic treatment, and parenteral nutrition are other factors which were demonstrated to delay cord separation. In the previous studies including greater premature infants, the time of separation of the umbilical cord was shown to vary 2 to 3 weeks. We aimed to determine the cord separation time and associated factors in very-low-birth-weight (VLBW) infants. STUDY DESIGN In this retrospective study, VLBW infants (birth weight [BW] <1,500 g, gestational age [GA] < 32 weeks) without umbilical catheterization were included. Specific cord care was not applied. The cord separation time, gender, mode of delivery, presence of sepsis, duration of antibiotic treatment, serum free thyroxine, free triiodothyronine (FT3), thyroid-stimulating hormone, lowest leukocyte, polymorphonuclear leukocytes (PMNLs), and platelet counts were recorded. RESULTS The study included 130 infants (GA: 29 ± 2 weeks, BW: 1,196 ± 243 g). Mean cord separation time was 14 ± 5 days, while 95th percentile was 22nd day of life. A positive correlation was demonstrated between duration of antibiotic treatment and cord separation time (p < 0.001, r: 0.505). Cord separation time did not differ regarding gender or mode of delivery. Corrected leukocyte count (p = 0.031, r: -0.190) and PMNL count (p = 0.022, r: -0.201), and serum FT3 level (p = 0.003, r: -0.261) were found to be negatively correlated with cord separation time. The cord separation time was found to be delayed in the presence of sepsis (with sepsis: 18 ± 7 days and without sepsis: 13 ± 3 days; p = 0.008). Sepsis was found to delay the cord separation time beyond second week (odds ratio = 6.30 [95% confidence interval: 2.37-15.62], p < 0.001). CONCLUSION The 95th percentile for cord separation time was 22nd day. Sepsis might be either the reason or the consequence of delayed cord detachment. The exact contribution of low serum FT3 levels to the process of cord separation should be investigated in further studies. KEY POINTS · Mean cord separation time was 14 ± 5 days, while 95th percentile was 22nd day, in VLBW infants.. · Sepsis was found to delay the cord separation time by sixfold beyond second week.. · Serum free triiodothyronine level was negatively correlated with cord separation time..
Collapse
Affiliation(s)
- Sezin Unal
- Department of Neonatology, Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Nihal Demirel
- Division of Neonatology, Department of Pediatrics, Yildirim Beyazit University Hospital, Ankara, Turkey
| | - Zehra Arslan
- Department of Neonatology, Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Betül Tokgoz-Cuni
- Department of Neonatology, Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Dilek Ulubas-Isik
- Department of Neonatology, Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Ahmet Yagmur Bas
- Division of Neonatology, Department of Pediatrics, Yildirim Beyazit University Hospital, Ankara, Turkey
| |
Collapse
|
32
|
Beudeker CR, Vijlbrief DC, van Montfrans J, Rooijakkers SH, van der Flier M. Neonatal sepsis and transient immunodeficiency: Potential for novel immunoglobulin therapies? Front Immunol 2022; 13:1016877. [PMID: 36330515 PMCID: PMC9623314 DOI: 10.3389/fimmu.2022.1016877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/04/2022] [Indexed: 10/30/2023] Open
Abstract
Neonates, especially preterm neonates, have the highest risk of sepsis of all age groups. Transient immaturity of the neonatal immune system is an important risk factor. Neonates suffer from hypogammaglobulinemia as nor IgA nor IgM is transferred over the placenta and IgG is only transferred over the placenta late in gestation. In addition, neutrophil numbers and complement function are also decreased. This mini-review focuses on strategies to improve neonatal host-defense. Both clinical and preclinical studies have attempted to boost neonatal immunity to lower the incidence of sepsis and improve outcome. Recent advances in the development of (monoclonal) antibodies show promising results in preclinical studies but have yet to be tested in clinical trials. Strategies to increase complement activity seem efficient in vitro but potential disadvantages such as hyperinflammation have held back further clinical development. Increase of neutrophil numbers has been tested extensively in clinical trials but failed to show improvement in mortality. Future research should focus on clinical applicability of promising new prevention strategies for neonatal sepsis.
Collapse
Affiliation(s)
- Coco R. Beudeker
- Department of Pediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel C. Vijlbrief
- Department of Neonatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joris M. van Montfrans
- Department of Pediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Suzan H.M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Michiel van der Flier
- Department of Pediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
33
|
Chen Y, Huang J, Guo Z, Zhu Z, Shao Y, Li L, Yang Y, Yu Y, Liu L, Sun B. Primitive genotypic characteristics in umbilical cord neutrophils identified by single-cell transcriptome profiling and functional prediction. Front Immunol 2022; 13:970909. [PMID: 36105817 PMCID: PMC9464600 DOI: 10.3389/fimmu.2022.970909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
The function and heterogeneity of neutrophils in neonatal umbilical cord blood (UCB) have not been characterized. In this study, we analyzed the neutrophils in UCB and healthy adults using single-cell RNA sequencing analysis for the first time. We found that neutrophils divided into six subpopulations (G2, G3, G4, G5a, G5b, and G5c) with different marker genes and different functions under homeostasis. Compared with healthy adults, neutrophils of UCB were more naïve and have more obvious degranulation and activation functions. Moreover, we found significant differences in the amount and function of G5b cells between healthy adults and UCB. The amount of G5b group in UCB was lower, but it has more degranulation, secretion and activation functions. In addition, we noted a new subset of G5c labeled by CD52, which almost did not exist in UCB. Besides, its differential genes were enriched in terms such as protein synthesis and mRNA transcription. Furthermore, uncharacteristic transcription factors ZNF-276, ZNF-319 and ZNF-354A were identified in our study. In summary, we first examined the heterogeneity and functional diversity of neutrophils in UCB, and these data provided new insights into the mechanism of neutrophil-mediated diseases of neonates and the wider use of neutrophils in UCB.
Collapse
Affiliation(s)
- Yi Chen
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Burns and Plastic Surgery, Affiliated Huaian No.1 People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Jiamin Huang
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zaiwen Guo
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhechen Zhu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Linbin Li
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yunxi Yang
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yanzhen Yu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lu Liu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Bingwei Sun,
| |
Collapse
|
34
|
Moise KJ, Oepkes D, Lopriore E, Bredius RGM. Targeting neonatal Fc receptor: potential clinical applications in pregnancy. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 60:167-175. [PMID: 35229965 DOI: 10.1002/uog.24891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
The neonatal Fc receptor (FcRn) plays an important role in the transfer of the immunoglobulin G isotype (IgG) from the mother to the fetus. FcRn expressed on endothelial cells also binds to IgG and albumin, regulating the circulating half-lives of these proteins. Alloimmune and autoimmune IgG antibodies have been implicated in various perinatal immune-mediated diseases. FcRn-mediated placental transfer of pathogenic antibodies can result in cell and tissue injury in the fetus and neonate, with devastating outcomes. Thus, blockade of FcRn may be an effective treatment strategy in managing these conditions and could additionally reduce the concentration of pathogenic antibodies in the maternal circulation by preventing IgG recycling. In this review, we discuss the biology of FcRn, the rationale and considerations for development of FcRn-blocking agents, and their potential clinical applications in various perinatal immune-mediated diseases. © 2022 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- K J Moise
- Department of Women's Health, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - D Oepkes
- Department of Obstetrics and Fetal Therapy, Leiden University Medical Center, Leiden, The Netherlands
| | - E Lopriore
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R G M Bredius
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
35
|
de Vor L, Beudeker CR, Flier A, Scheepmaker LM, Aerts PC, Vijlbrief DC, Bekker MN, Beurskens FJ, van Kessel KPM, de Haas CJC, Rooijakkers SHM, van der Flier M. Monoclonal antibodies effectively potentiate complement activation and phagocytosis of Staphylococcus epidermidis in neonatal human plasma. Front Immunol 2022; 13:933251. [PMID: 35967335 PMCID: PMC9372458 DOI: 10.3389/fimmu.2022.933251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
Central line associated bloodstream infections (CLABSI) with Staphylococcus epidermidis are a major cause of morbidity in neonates, who have an increased risk of infection because of their immature immune system. As especially preterm neonates suffer from antibody deficiency, clinical studies into preventive therapies have thus far focused on antibody supplementation with pooled intravenous immunoglobulins from healthy donors (IVIG) but with little success. Here we study the potential of monoclonal antibodies (mAbs) against S. epidermidis to induce phagocytic killing by human neutrophils. Nine different mAbs recognizing Staphylococcal surface components were cloned and expressed as human IgG1s. In binding assays, clones rF1, CR5133 and CR6453 showed the strongest binding to S. epidermidis ATCC14990 and CR5133 and CR6453 bound the majority of clinical isolates from neonatal sepsis (19 out of 20). To study the immune-activating potential of rF1, CR5133 and CR6453, bacteria were opsonized with mAbs in the presence or absence of complement. We observed that activation of the complement system is essential to induce efficient phagocytosis of S. epidermidis. Complement activation and phagocytic killing could be enhanced by Fc-mutations that improve IgG1 hexamerization on cellular surfaces. Finally, we studied the ability of the mAbs to activate complement in r-Hirudin neonatal plasma conditions. We show that classical pathway complement activity in plasma isolated from neonatal cord blood is comparable to adult levels. Furthermore, mAbs could greatly enhance phagocytosis of S. epidermidis in neonatal plasma. Altogether, our findings provide insights that are crucial for optimizing anti-S. epidermidis mAbs as prophylactic agents for neonatal CLABSI.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Coco R. Beudeker
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anne Flier
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lisette M. Scheepmaker
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel C. Vijlbrief
- Department of Neonatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mireille N. Bekker
- Department of Obstetrics, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Kok P. M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Carla J. C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Michiel van der Flier
- Department of Paediatric Infectious Diseases and Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
36
|
Wang L, Rajavel M, Wu CW, Zhang C, Poindexter M, Fulgar C, Mar T, Singh J, Dhillon JK, Zhang J, Yuan Y, Abarca R, Li W, Pinkerton KE. Effects of life-stage and passive tobacco smoke exposure on pulmonary innate immunity and influenza infection in mice. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:439-456. [PMID: 35139765 PMCID: PMC8976777 DOI: 10.1080/15287394.2022.2032518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Limited data are available on the effects of perinatal environmental tobacco smoke (ETS) exposure for early childhood influenza infection. The aim of the present study was to examine whether perinatal versus adult ETS exposure might provoke more severe systemic and pulmonary innate immune responses in mice inoculated with influenza A/Puerto Rico/8/34 virus (IAV) compared to phosphate-buffered saline (PBS). BALB/c mice were exposed to filtered air (FA) or ETS for 6 weeks during the perinatal or adult period of life. Immediately following the final exposure, mice were intranasally inoculated with IAV or PBS. Significant inflammatory effects were observed in bronchoalveolar lavage fluid of neonates inoculated with IAV (FA+IAV or ETS+IAV) compared to PBS (ETS+PBS or FA+PBS), and in the lung parenchyma of neonates administered ETS+IAV versus FA+IAV. Type I and III interferons were also elevated in the spleens of neonates, but not adults with ETS+IAV versus FA+IAV exposure. Both IAV-inoculated neonate groups exhibited significantly more CD4 T cells and increasing numbers of CD8 and CD25 T cells in lungs relative to their adult counterparts. Taken together, these results suggest perinatal ETS exposure induces an exaggerated innate immune response, which may overwhelm protective anti-inflammatory defenses against IAV, and enhances severity of infection at early life stages (e.g., in infants and young children).
Collapse
Affiliation(s)
- Lei Wang
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Maya Rajavel
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Chuanzhen Zhang
- Center for Health and the Environment, University of California, Davis, CA, USA
- Department of Gastroenterology, the First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Morgan Poindexter
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Ciara Fulgar
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Tiffany Mar
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Jasmine Singh
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Jaspreet K. Dhillon
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Jingjing Zhang
- Center for Health and the Environment, University of California, Davis, CA, USA
- Western China School of Public Health Department of Occupational and Environmental Health Sichuan University, Chengdu, China
| | - Yinyu Yuan
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Radek Abarca
- Center for Health and the Environment, University of California, Davis, CA, USA
| | - Wei Li
- School of Control Science and Engineering, Shandong University, Jinan, Shandong 250014, China
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, CA, USA
- Department of Pediatrics, University of California, Davis, CA, USA
| |
Collapse
|
37
|
Abstract
BACKGROUND Extreme immature infants are at an increased risk of fungal infection due to immaturity of the skin barrier and the immune system. Besides Candida infections, in particular, Aspergillus may cause life-threatening diseases in preterm infants. Frequently, Aspergillus primarily affects the skin and may cause extensive damage. METHODS We searched our hospital database for fungal infections other than Candida in preterm infants treated between 2015 and 2020 at our level III neonatal intensive care unit of the University Hospital of Cologne. RESULTS In total, 13 preterm infants were identified. Of these, 11 had cutaneous Aspergillosis, one infant had severe enterocolitis caused by Aspergillus and Rhizopus and one had invasive intraabdominal Trichosporon mucoides infection. All infants were born <24 weeks of gestation, were delivered due to premature labor or chorioamnionitis, and had received prenatal steroids and/or hydrocortisone. Voriconazole and liposomal Amphotericin B were first-line treatments and the length of treatment varied between 3 and 148 days. Two infants died associated with severe infection. Liver toxicity was observed in six infants treated with Voriconazole. Therapeutic drug management for Voriconazole was performed in four infants. Target levels were not achieved by the doses that are recommended. CONCLUSIONS Rare fungal infections, predominantly cutaneous Aspergillosis affects the most immature preterm infants and may cause severe disease. Treatment with Voriconazole has a high rate of liver toxicity and target levels are difficult to achieve in extremely immature infants.
Collapse
|
38
|
Jackson CM, Demmert M, Mukherjee S, Isaacs T, Thompson R, Chastain C, Gray J, Senthamaraikannan P, Presicce P, Chetal K, Salomonis N, Miller LA, Jobe AH, Kallapur SG, Zacharias WJ, Lewkowich IP, Deshmukh H, Chougnet CA. A potent myeloid response is rapidly activated in the lungs of premature Rhesus macaques exposed to intra-uterine inflammation. Mucosal Immunol 2022; 15:730-744. [PMID: 35314757 PMCID: PMC9259482 DOI: 10.1038/s41385-022-00495-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023]
Abstract
Up to 40% of preterm births are associated with histological chorioamnionitis (HCA), which leads to elevated levels of pro-inflammatory mediators and microbial products in the amniotic fluid, which come in contact with fetal lungs. Yet, fetal pulmonary immune responses to such exposure remain poorly characterized. To address this gap, we used our established HCA model, in which pregnant Rhesus macaques receive intraamniotic (IA) saline or LPS. IA LPS induced a potent and rapid myeloid cell response in fetal lungs, dominated by neutrophils and monocytes/macrophages. Infiltrating and resident myeloid cells exhibited transcriptional profiles consistent with exposure to TLR ligands, as well as cytokines, notably IL-1 and TNFα. Although simultaneous, in vivo blockade of IL-1 and TNFα signaling did not prevent the inflammatory cell recruitment, it blunted the lung overall inflammatory state reducing communication between, and activation of, infiltrating immune cells. Our data indicate that the fetal innate immune system can mount a rapid multi-faceted pulmonary immune response to in utero exposure to inflammation. These data provide mechanistic insights into the association between HCA and the postnatal lung morbidities of the premature infant and highlight therapeutic potential of inflammatory blockade in the fetus.
Collapse
Affiliation(s)
- Courtney M Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Martin Demmert
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Pediatrics, Institute for Systemic Inflammation Research, University of Lϋbeck, Lϋbeck, Germany.
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Travis Isaacs
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ravyn Thompson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chase Chastain
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jerilyn Gray
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paranth Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center, University of California Davis, Davis, CA, USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - William J Zacharias
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ian P Lewkowich
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hitesh Deshmukh
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
39
|
Peripheral immune cells and perinatal brain injury: a double-edged sword? Pediatr Res 2022; 91:392-403. [PMID: 34750522 PMCID: PMC8816729 DOI: 10.1038/s41390-021-01818-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023]
Abstract
Perinatal brain injury is the leading cause of neurological mortality and morbidity in childhood ranging from motor and cognitive impairment to behavioural and neuropsychiatric disorders. Various noxious stimuli, including perinatal inflammation, chronic and acute hypoxia, hyperoxia, stress and drug exposure contribute to the pathogenesis. Among a variety of pathological phenomena, the unique developing immune system plays an important role in the understanding of mechanisms of injury to the immature brain. Neuroinflammation following a perinatal insult largely contributes to evolution of damage to resident brain cells, but may also be beneficial for repair activities. The present review will focus on the role of peripheral immune cells and discuss processes involved in neuroinflammation under two frequent perinatal conditions, systemic infection/inflammation associated with encephalopathy of prematurity (EoP) and hypoxia/ischaemia in the context of neonatal encephalopathy (NE) and stroke at term. Different immune cell subsets in perinatal brain injury including their infiltration routes will be reviewed and critical aspects such as sex differences and maturational stage will be discussed. Interactions with existing regenerative therapies such as stem cells and also potentials to develop novel immunomodulatory targets are considered. IMPACT: Comprehensive summary of current knowledge on the role of different immune cell subsets in perinatal brain injury including discussion of critical aspects to be considered for development of immunomodulatory therapies.
Collapse
|
40
|
Xing Z, Zhen T, Jie F, Jie Y, Shiqi L, Kaiyi Z, Zhicui O, Mingyan H. Early Toll-like receptor 4 inhibition improves immune dysfunction in the hippocampus after hypoxic-ischemic brain damage. Int J Med Sci 2022; 19:142-151. [PMID: 34975308 PMCID: PMC8692118 DOI: 10.7150/ijms.66494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Toll-like receptor 4 (TLR4) is implicated in neonatal hypoxic-ischemic brain damage (HIBD), but the underlying mechanism is unclear. Hypothesis: We hypothesized that TLR4 mediates brain damage after hypoxic ischemia (HI) by inducing abnormal neuroimmune responses, including activation of immune cells and expression disorder of immune factors, while early inhibition of TLR4 can alleviate the neuroimmune dysfunction. Method: Postnatal day 7 rats were randomized into control, HI, and HI+TAK-242 (TAK-242) groups. The HIBD model was developed using the Rice-Vannucci method (the left side was the ipsilateral side of HI). TAK-242 (0.5 mg/kg) was given to rat pups in the TAK-242 group at 30 min before modeling. Immunofluorescence, immunohistochemistry, and western blotting were used to determine the TLR4 expression; the number of Iba-1+, GFAP+, CD161+, MPO+, and CD3+ cells; ICAM-1 and C3a expression; and interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-10 expression in the hippocampal CA1 region. Result: Significantly increased TLR4 expression was observed in the left hippocampus, and was alleviated by TAK-242. The significant increases in Iba-1+, MPO+, and CD161+ cells at 24 h and 7 days after HI and in GFAP+ and CD3+ T cells at 7 days after HI were also counteracted by TAK-242, but no significant differences were observed among groups at 24 h after HI. ICAM-1 expression increased 24 h after HI, while C3a expression decreased; TAK-242 also alleviated these changes. TNF-α and IL-1β expression increased, while IL-10 expression decreased at 24 h and 7 days after HI; TAK-242 counteracted the increased TNF-α and IL-1β expression at 24 h and the changes in IL-1β and IL-10 at 7 days, but induced no significant differences in IL-10 expression at 24 h and TNF-α expression at 7 days. Conclusion: Early TLR4 inhibition can alleviate hippocampal immune dysfunction after neonatal HIBD.
Collapse
Affiliation(s)
- Zhu Xing
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Tang Zhen
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China.,Department of Pediatrics, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013 China
| | - Fan Jie
- Department of Neonatology, East Hospital of Shaoyang Central Hospital, Shaoyang, Hunan, 422000 China
| | - Yu Jie
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Liu Shiqi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Zhu Kaiyi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - OuYang Zhicui
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China
| | - Hei Mingyan
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| |
Collapse
|
41
|
Rayasam A, Jullienne A, Chumak T, Faustino J, Szu J, Hamer M, Ek CJ, Mallard C, Obenaus A, Vexler ZS. Viral mimetic triggers cerebral arteriopathy in juvenile brain via neutrophil elastase and NETosis. J Cereb Blood Flow Metab 2021; 41:3171-3186. [PMID: 34293939 PMCID: PMC8669290 DOI: 10.1177/0271678x211032737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Stroke is among the top ten causes of death in children but has received disproportionally little attention. Cerebral arteriopathies account for up to 80% of childhood arterial ischemic stroke (CAIS) cases and are strongly predictive of CAIS recurrence and poorer outcomes. The underlying mechanisms of sensitization of neurovasculature by viral infection are undefined. In the first age-appropriate model for childhood arteriopathy-by administration of viral mimetic TLR3-agonist Polyinosinic:polycytidylic acid (Poly-IC) in juvenile mice-we identified a key role of the TLR3-neutrophil axis in disrupting the structural-functional integrity of the blood-brain barrier (BBB) and distorting the developing neurovascular architecture and vascular networks. First, using an array of in-vivo/post-vivo vascular imaging, genetic, enzymatic and pharmacological approaches, we report marked Poly-IC-mediated extravascular leakage of albumin (66kDa) and of a small molecule DiI (∼934Da) and disrupted tight junctions. Poly-IC also enhanced the neuroinflammatory milieu, promoted neutrophil recruitment, profoundly upregulated neutrophil elastase (NE), and induced neutrophil extracellular trap formation (NETosis). Finally, we show that functional BBB disturbances, NETosis and neuroinflammation are markedly attenuated by pharmacological inhibition of NE (Sivelestat). Altogether, these data reveal NE/NETosis as a novel therapeutic target for viral-induced cerebral arteriopathies in children.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Neurology, University California San Francisco, San Francisco, CA, USA
| | - Amandine Jullienne
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Tetyana Chumak
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Faustino
- Department of Neurology, University California San Francisco, San Francisco, CA, USA
| | - Jenny Szu
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Mary Hamer
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - C Joakim Ek
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, San Francisco, CA, USA
| |
Collapse
|
42
|
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
|
43
|
Campbell RA, Campbell HD, Bircher JS, de Araujo CV, Denorme F, Crandell JL, Rustad JL, Monts J, Cody MJ, Kosaka Y, Yost CC. Placental HTRA1 cleaves α1-antitrypsin to generate a NET-inhibitory peptide. Blood 2021; 138:977-988. [PMID: 34192300 PMCID: PMC9069473 DOI: 10.1182/blood.2020009021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/30/2021] [Accepted: 06/13/2021] [Indexed: 11/20/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are important components of innate immunity. Neonatal neutrophils (polymorphonuclear leukocytes [PMNs]) fail to form NETs due to circulating NET-inhibitory peptides (NIPs), cleavage fragments of α1-antitrypsin (A1AT). How fetal and neonatal blood NIPs are generated remains unknown, however. The placenta expresses high-temperature requirement serine protease A1 (HTRA1) during fetal development, which can cleave A1AT. We hypothesized that placentally expressed HTRA1 regulates the formation of NIPs and that NET competency changed in PMNs isolated from neonatal HTRA1 knockout mice (HTRA1-/-). We found that umbilical cord blood plasma has elevated HTRA1 levels compared with adult plasma and that recombinant and placenta-eluted HTRA1 cleaves A1AT to generate an A1AT cleavage fragment (A1ATM383S-CF) of molecular weight similar to previously identified NIPs that block NET formation by adult neutrophils. We showed that neonatal mouse pup plasma contains A1AT fragments that inhibit NET formation by PMNs isolated from adult mice, indicating that NIP generation during gestation is conserved across species. Lipopolysaccharide-stimulated PMNs isolated from HTRA1+/+ littermate control pups exhibit delayed NET formation after birth. However, plasma from HTRA1-/- pups had no detectable NIPs, and PMNs from HTRA1-/- pups became NET competent earlier after birth compared with HTRA1+/+ littermate controls. Finally, in the cecal slurry model of neonatal sepsis, A1ATM383S-CF improved survival in C57BL/6 pups by preventing pathogenic NET formation. Our data indicate that placentally expressed HTRA1 is a serine protease that cleaves A1AT in utero to generate NIPs that regulate NET formation by human and mouse PMNs.
Collapse
Affiliation(s)
- Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Internal Medicine
| | | | | | | | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Jacob L Crandell
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - John L Rustad
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Josh Monts
- Flow Cytometry Core, University of Utah, Salt Lake City, UT
| | - Mark J Cody
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
| | - Christian C Yost
- University of Utah Molecular Medicine Program, Salt Lake City, UT; and
- Department of Pediatrics, and
| |
Collapse
|
44
|
Provencher A, Katavolos P. Brief Synopsis: "Instruction Manual for Juvenile Clinical Pathology". Toxicol Pathol 2021; 49:1389-1392. [PMID: 34431401 DOI: 10.1177/01926233211041338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This symposium synopsis summarizes key points discussed related to clinical pathology data interpretation for reproduction and juvenile toxicology studies. In pregnant and growing animals, several changes in clinical pathology parameters linked to growth/maturation of organ and physiological functions can occur, and understanding these changes is important to enable accurate interpretation of clinical pathology data. A brief overview of the general approach to clinical pathology data analysis according to contemporary practices is provided, followed by a discussion focused specifically on reproductive and juvenile clinical pathology. In this context, the approach to recognize and differentiate changes that may be related to pregnancy and growth as opposed to those that may be related to test article effects is highlighted.
Collapse
Affiliation(s)
- Anne Provencher
- Global Safety Assessment, 70294Charles River Laboratories, Sherbrooke, Quebec, Canada
| | - Paula Katavolos
- Nonclinical Safety, 137687Bristol Myers Squibb, New Brunswick, NJ, USA
| |
Collapse
|
45
|
Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, Linnekin T, Kulkarni V, Lu R, Slein MD, Luedemann C, Marquette M, March S, Weiner J, Gregory S, Coccia M, Flores-Garcia Y, Zavala F, Ackerman ME, Bergmann-Leitner E, Hendriks J, Sadoff J, Dutta S, Bhatia SN, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med 2021; 12:12/553/eabb4757. [PMID: 32718991 DOI: 10.1126/scitranslmed.abb4757] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
Vaccine development has the potential to be accelerated by coupling tools such as systems immunology analyses and controlled human infection models to define the protective efficacy of prospective immunogens without expensive and slow phase 2b/3 vaccine studies. Among human challenge models, controlled human malaria infection trials have long been used to evaluate candidate vaccines, and RTS,S/AS01 is the most advanced malaria vaccine candidate, reproducibly demonstrating 40 to 80% protection in human challenge studies in malaria-naïve individuals. Although antibodies are critical for protection after RTS,S/AS01 vaccination, antibody concentrations are inconsistently associated with protection across studies, and the precise mechanism(s) by which vaccine-induced antibodies provide protection remains enigmatic. Using a comprehensive systems serological profiling platform, the humoral correlates of protection against malaria were identified and validated across multiple challenge studies. Rather than antibody concentration, qualitative functional humoral features robustly predicted protection from infection across vaccine regimens. Despite the functional diversity of vaccine-induced immune responses across additional RTS,S/AS01 vaccine studies, the same antibody features, antibody-mediated phagocytosis and engagement of Fc gamma receptor 3A (FCGR3A), were able to predict protection across two additional human challenge studies. Functional validation using monoclonal antibodies confirmed the protective role of Fc-mediated antibody functions in restricting parasite infection both in vitro and in vivo, suggesting that these correlates may mechanistically contribute to parasite restriction and can be used to guide the rational design of an improved vaccine against malaria.
Collapse
Affiliation(s)
- Todd J Suscovich
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Jishnu Das
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Allison R Demas
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Crain
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Caitlyn H Linde
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Ashlin Michell
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Harini Natarajan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Claudia Arevalo
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Broge
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Linnekin
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Viraj Kulkarni
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Richard Lu
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew D Slein
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Meghan Marquette
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Elke Bergmann-Leitner
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jenny Hendriks
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Jerald Sadoff
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Broad Institute, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
46
|
Exploring Clinically-Relevant Experimental Models of Neonatal Shock and Necrotizing Enterocolitis. Shock 2021; 53:596-604. [PMID: 31977960 DOI: 10.1097/shk.0000000000001507] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neonatal shock and necrotizing enterocolitis (NEC) are leading causes of morbidity and mortality in premature infants. NEC is a life-threatening gastrointestinal illness, the precise etiology of which is not well understood, but is characterized by an immaturity of the intestinal barrier, altered function of the adaptive immune system, and intestinal dysbiosis. The complexities of NEC and shock in the neonatal population necessitate relevant clinical modeling using newborn animals that mimic the disease in human neonates to better elucidate the pathogenesis and provide an opportunity for the discovery of potential therapeutics. A wide variety of animal species-including rats, mice, piglets, and primates-have been used in developing experimental models of neonatal diseases such as NEC and shock. This review aims to highlight the immunologic differences in neonates compared with adults and provide an assessment of the advantages and drawbacks of established animal models of both NEC and shock using enteral or intraperitoneal induction of bacterial pathogens. The selection of a model has benefits unique to each type of animal species and provides individual opportunities for the development of targeted therapies. This review discusses the clinical and physiologic relevance of animal models and the insight they contribute to the complexities of the specific neonatal diseases: NEC and shock.
Collapse
|
47
|
Vrachnis N, Zygouris D, Vrachnis D, Roussos N, Loukas N, Antonakopoulos N, Paltoglou G, Barbounaki S, Valsamakis G, Iliodromiti Z. Perinatal Inflammation: Could Partial Blocking of Cell Adhesion Molecule Function Be a Solution? CHILDREN-BASEL 2021; 8:children8050380. [PMID: 34065912 PMCID: PMC8150343 DOI: 10.3390/children8050380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 11/16/2022]
Abstract
In spite of the great advances made in recent years in prenatal and perinatal medicine, inflammation can still frequently result in injury to vital organs and often constitutes a major cause of morbidity. It is today well established that in neonates—though vulnerability to infection among neonates is triggered by functional impairments in leukocyte adhesion—the decreased expression of cell adhesion molecules also decreases the inflammatory response. It is also clear that the cell adhesion molecules, namely, the integrins, selectins, and the immunoglobulin (Ig) gene super family, all play a crucial role in the inflammatory cascade. Thus, by consolidating our knowledge concerning the actions of these vital cell adhesion molecules during the prenatal period as well as regarding the genetic deficiencies of these molecules, notably leukocyte adhesion deficiency (LAD) I, II, and III, which can provoke severe clinical symptoms throughout the first year of life, it is anticipated that intervention involving blocking the function of cell adhesion molecules in neonatal leukocytes has the potential to constitute an effective therapeutic approach for inflammation. A promising perspective is the potential use of antibody therapy in preterm and term infants with perinatal inflammation and infection focusing on cases in which LAD is involved, while a further important scientific advance related to this issue could be the combination of small peptides aimed at the inhibition of cellular adhesion.
Collapse
Affiliation(s)
- Nikolaos Vrachnis
- Third Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 11526 Athens, Greece;
- Vascular Biology, Molecular and Clinical Sciences Research Institute, St George’s University of London, London SW17 0RE, UK
- Research Centre in Obstetrics and Gynecology, Hellenic Society of Obstetric and Gynecologic Emergency, 11526 Athens, Greece; (D.Z.); (N.R.)
- Correspondence: ; Tel.: +30-2107777442
| | - Dimitrios Zygouris
- Research Centre in Obstetrics and Gynecology, Hellenic Society of Obstetric and Gynecologic Emergency, 11526 Athens, Greece; (D.Z.); (N.R.)
| | - Dionysios Vrachnis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Alexandra Hospital, 11526 Athens, Greece;
| | - Nikolaos Roussos
- Research Centre in Obstetrics and Gynecology, Hellenic Society of Obstetric and Gynecologic Emergency, 11526 Athens, Greece; (D.Z.); (N.R.)
| | - Nikolaos Loukas
- Department of Gynecology, General Hospital of Athens “G. Gennimatas”, 11527 Athens, Greece;
| | - Nikolaos Antonakopoulos
- Third Department of Obstetrics and Gynecology, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 11526 Athens, Greece;
| | - Georgios Paltoglou
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, School of Medicine, National and Kapodistrian University of Athens, Aretaieion Hospital, 11526 Athens, Greece; (G.P.); (G.V.)
| | | | - Georgios Valsamakis
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, School of Medicine, National and Kapodistrian University of Athens, Aretaieion Hospital, 11526 Athens, Greece; (G.P.); (G.V.)
| | - Zoi Iliodromiti
- Department of Neonatology, School of Medicine, National and Kapodistrian University of Athens, Aretaieio Hospital, 11526 Athens, Greece;
| |
Collapse
|
48
|
Rabi FA, Al Zoubi MS, Al‐Iede MM, Kasasbeh G, Badran EF. Coronaviruses in children: A review of potential mechanisms of childhood protection. Acta Paediatr 2021; 110:765-772. [PMID: 33247469 PMCID: PMC7753280 DOI: 10.1111/apa.15691] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/28/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022]
Abstract
Aim The 2019 coronavirus disease (COVID‐19) has spread worldwide and the number of cases continues to rise exponentially. Epidemiologic reports indicate that severity of illness increases with age. However, the reasons behind the relative protection of children and infants are unclear. Whether the rationale is host‐related or virus dependent is important to determine since the latter could change with viral mutations. We review factors that could affect the susceptibility of children to the novel coronavirus. Methods We search publications indexed on PUBMED. Results Descriptions of the pathophysiology of current and previous coronavirus infections suggest several viral targets and immunomodulatory pathways affecting the severity of illness. There is limited evidence to suggest age‐variability of viral cell receptors and transmembrane co‐factors required for coronavirus entry and replication. However, the ensuing cytokine storm and the effect of higher melatonin in children are age‐dependent and could explain decreased disease variability in children. Conclusion We believe that current evidence suggests host factors can play a role in disease severity in children and thus may remain protective despite potential virus mutation in the future. However, we recognise and discuss avenues of future research that can further illuminate the reasons children are protected from severe COVID‐19 illness.
Collapse
Affiliation(s)
- Firas A. Rabi
- Department of Clinical Sciences Faculty of Medicine Yarmouk University Irbid Jordan
| | - Mazhar S. Al Zoubi
- Department of Basic Medical Sciences Faculty of Medicine Yarmouk University Irbid Jordan
| | | | | | - Eman Farouk Badran
- Department of Pediatrics Faculty of Medicine Jordan University Amman Jordan
| |
Collapse
|
49
|
Turturice BA, Theorell J, Koenig MD, Tussing-Humphreys L, Gold DR, Litonjua AA, Oken E, Rifas-Shiman SL, Perkins DL, Finn PW. Perinatal granulopoiesis and risk of pediatric asthma. eLife 2021; 10:e63745. [PMID: 33565964 PMCID: PMC7889076 DOI: 10.7554/elife.63745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022] Open
Abstract
There are perinatal characteristics, such as gestational age, reproducibly associated with the risk for pediatric asthma. Identification of biologic processes influenced by these characteristics could facilitate risk stratification or new therapeutic targets. We hypothesized that transcriptional changes associated with multiple epidemiologic risk factors would be mediators of pediatric asthma risk. Using publicly available transcriptomic data from cord blood mononuclear cells, transcription of genes involved in myeloid differentiation was observed to be inversely associated with a pediatric asthma risk stratification based on multiple perinatal risk factors. This gene signature was validated in an independent prospective cohort and was specifically associated with genes localizing to neutrophil-specific granules. Further validation demonstrated that umbilical cord blood serum concentration of PGLYRP-1, a specific granule protein, was inversely associated with mid-childhood current asthma and early-teen FEV1/FVCx100. Thus, neutrophil-specific granule abundance at birth predicts risk for pediatric asthma and pulmonary function in adolescence.
Collapse
Affiliation(s)
- Benjamin A Turturice
- Department of Microbiology and Immunology, University of IllinoisChicagoUnited States
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of IllinoisChicagoUnited States
| | - Juliana Theorell
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of IllinoisChicagoUnited States
| | - Mary Dawn Koenig
- Department of Women, Children and Family Health Science, College of Nursing, University of IllinoisChicagoUnited States
| | | | - Diane R Gold
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Environmental Health, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Augusto A Litonjua
- Division of Pulmonary Medicine, Department of Pediatrics, University of RochesterRochesterUnited States
| | - Emily Oken
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care InstituteBostonUnited States
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care InstituteBostonUnited States
| | - David L Perkins
- Department of Medicine, Division of Nephrology, University of IllinoisChicagoUnited States
- Department of Bioengineering, University of IllinoisChicagoUnited States
| | - Patricia W Finn
- Department of Microbiology and Immunology, University of IllinoisChicagoUnited States
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of IllinoisChicagoUnited States
- Department of Bioengineering, University of IllinoisChicagoUnited States
| |
Collapse
|
50
|
Mülling K, Fischer AJ, Siakaeva E, Richter M, Bordbari S, Spyra I, Köster C, Hermann DM, Gunzer M, Felderhoff-Müser U, Bendix I, Jablonska J, Herz J. Neutrophil dynamics, plasticity and function in acute neurodegeneration following neonatal hypoxia-ischemia. Brain Behav Immun 2021; 92:234-244. [PMID: 33333168 DOI: 10.1016/j.bbi.2020.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Neonatal encephalopathy following hypoxia-ischemia (HI) is a major cause of long-term morbidity and mortality in children. Even though HI-induced neuroinflammation, involving infiltration of peripheral immune cells into the CNS has been associated with disease pathogenesis, the specific role of neutrophils is highly debated. Due to immaturity of the neonatal immune system, it has been assumed that neutrophils are less clinically relevant in neonatal HI-induced brain injury. In the present study, we demonstrate that neutrophils are rapidly activated in the neonatal brain after exposure to experimental HI, revealed by an enhanced proportion of CD86+ cells and an increased expression of CD11b compared to splenic and blood neutrophils. Furthermore, production of reactive oxygen species and the proportion of hyperactivated/aged (CXCR4+CD62L-) cells was enhanced in brain compared to peripheral neutrophils. Delayed neutrophil depletion, initiated 12 h after HI resulted in reduced cellular neurodegeneration, associated with reduced micro- and astroglial activation. In the present study, we uncovered a new complex switch of the phenotype in brain neutrophils, which may offer new possibilities for the development of selective therapeutic approaches by modulation of neutrophils in the early post-hypoxic disease phase.
Collapse
Affiliation(s)
- Kerstin Mülling
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexa Josephine Fischer
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Elena Siakaeva
- Department of Otorhinolaryngology, Translational Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mathis Richter
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sharareh Bordbari
- Department of Otorhinolaryngology, Translational Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ilona Spyra
- Department of Otorhinolaryngology, Translational Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Köster
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jadwiga Jablonska
- Department of Otorhinolaryngology, Translational Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK) partner site Düsseldorf/Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|