1
|
Chen CJ, Yi H, Stanley N. Conditional similarity triplets enable covariate-informed representations of single-cell data. BMC Bioinformatics 2025; 26:45. [PMID: 39924480 PMCID: PMC11807331 DOI: 10.1186/s12859-025-06069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Single-cell technologies enable comprehensive profiling of diverse immune cell-types through the measurement of multiple genes or proteins per individual cell. In order to translate immune signatures assayed from blood or tissue into powerful diagnostics, machine learning approaches are often employed to compute immunological summaries or per-sample featurizations, which can be used as inputs to models for outcomes of interest. Current supervised learning approaches for computing per-sample representations are trained only to accurately predict a single outcome and do not take into account relevant additional clinical features or covariates that are likely to also be measured for each sample. RESULTS Here, we introduce a novel approach for incorporating measured covariates in optimizing model parameters to ultimately specify per-sample encodings that accurately affect both immune signatures and additional clinical information. Our introduced method CytoCoSet is a set-based encoding method for learning per-sample featurizations, which formulates a loss function with an additional triplet term penalizing samples with similar covariates from having disparate embedding results in per-sample representations. CONCLUSIONS Overall, incorporating clinical covariates enables the learning of encodings for each individual sample that ultimately improve prediction of clinical outcome. This integration of information disparate more robust predictions of clinical phenotypes and holds significant potential for enhancing diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Chi-Jane Chen
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Haidong Yi
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Natalie Stanley
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
2
|
Mithal LB, Lancki N, Ling-Hu T, Goo YA, Otero S, Rhodes NJ, Cho BK, Grobman WA, Hultquist JF, Scholtens D, Mestan KK, Seed PC. Evolution of the umbilical cord blood proteome across gestational development. Sci Rep 2025; 15:2233. [PMID: 39825080 PMCID: PMC11742406 DOI: 10.1038/s41598-024-84446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025] Open
Abstract
Neonatal health is dependent on early risk stratification, diagnosis, and timely management of potentially devastating conditions, particularly in the setting of prematurity. Many of these conditions are poorly predicted in real-time by clinical data and current diagnostics. Umbilical cord blood may represent a novel source of molecular signatures that provides a window into the state of the fetus at birth. In this study, we comprehensively characterized the cord blood proteome of infants born between 25 to 42 weeks using untargeted mass spectrometry and functional enrichment analysis. We determined that the cord blood proteome at birth varies significantly across gestational development. Proteins that function in structural development and growth (e.g., extracellular matrix organization, lipid particle remodeling, and blood vessel development) are more abundant earlier in gestation. In later gestations, proteins with increased abundance are in immune response and inflammatory pathways, including complements and calcium-binding proteins. These data contribute to the knowledge of the physiologic state of neonates across gestational age, which is crucial to understand as we strive to best support postnatal development in preterm infants, determine mechanisms of pathology causing adverse health outcomes, and develop cord blood biomarkers to help tailor our diagnosis and therapeutics for critical neonatal conditions.
Collapse
Affiliation(s)
- Leena B Mithal
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Nicola Lancki
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ted Ling-Hu
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Ah Goo
- Mass Spectrometry Technology Access Center at McDonnell Genome Institute (MTAC@MGI), Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Sebastian Otero
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nathaniel J Rhodes
- Department of Pharmacy Practice, College of Pharmacy, Midwestern University, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Midwestern University, Downers Grove, IL, USA
- Department of Pharmacy, Northwestern Memorial Hospital, Chicago, IL, USA
| | - Byoung-Kyu Cho
- Mass Spectrometry Technology Access Center at McDonnell Genome Institute (MTAC@MGI), Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - William A Grobman
- Department of Obstetrics and Gynecology, Ohio State University, Columbus, OH, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise Scholtens
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karen K Mestan
- Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, CA, USA
| | - Patrick C Seed
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
van Duuren IC, van Hengel ORJ, Penders J, Duijts L, Smits HH, Tramper-Stranders GA. The developing immune system in preterm born infants: From contributor to potential solution for respiratory tract infections and wheezing. Allergy 2024; 79:2924-2942. [PMID: 39382056 DOI: 10.1111/all.16342] [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: 06/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Moderate-late preterm-born infants experience more frequent and severe respiratory tract infections and wheezing compared to term-born infants. Decreasing the risk on respiratory tract infections and wheezing in this group is vital to improve quality of life and reduce medical consumption during infancy, but also to reduce the risk on asthma and COPD later in life. Until now, moderate-late preterm infants are underrepresented in research and mechanisms underlying their morbidity are largely unknown, although they represent 80% of all preterm-born infants. In order to protect these infants effectively, it is essential to understand the role of the immune system in early life respiratory health and to identify strategies to optimize immune development and respiratory health. This review elaborates on risk factors and preventative measures concerning respiratory tract infections and wheezing in preterm-born infants, exploring their impact on the immune system and microbiome. Factors discussed are early life antibiotic use, birth mode, feeding type and living environment. Further, differences in adaptive and innate immune maturation between term and preterm infants are discussed, as well as differences in local immune reactions in the lungs. Finally, preventative strategies are being explored, including microbiota transplantation, immune modulation (through pre-, pro-, syn- and postbiotics, bacterial lysates, vaccinations, and monoclonal antibodies) and antibiotic prophylaxis.
Collapse
Affiliation(s)
- Inger C van Duuren
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Sophia Children's Hospital - Erasmus MC, Rotterdam, The Netherlands
| | - Oscar R J van Hengel
- Leiden University Center of Infectious Disease (LU-CID), Leiden, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Sophia Children's Hospital - Erasmus MC, Rotterdam, The Netherlands
| | - Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden, The Netherlands
| | - Gerdien A Tramper-Stranders
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Sophia Children's Hospital, ErasmusMC, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Chen Y, Wen Y, Zhao R, Zhu Y, Chen Z, Zhao C, Mu W. Human milk oligosaccharides in preventing food allergy: A review through gut microbiota and immune regulation. Int J Biol Macromol 2024; 278:134868. [PMID: 39163965 DOI: 10.1016/j.ijbiomac.2024.134868] [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: 04/30/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Food allergy (FA) has increasingly attracted global attention in past decades. However, the mechanism and effect of FA are complex and varied, rendering it hard to prevention and management. Most of the allergens identified so far are macromolecular proteins in food and may have potential cross-reactions. Human milk oligosaccharides (HMOs) have been regarded as an ideal nutrient component for infants, as they can enhance the immunomodulatory capacity to inhibit the progress of FA. HMOs may intervene in the development of allergies by modifying gut microbiota and increasing specific short-chain fatty acids levels. Additionally, HMOs could improve the intestinal permeability and directly or indirectly regulate the balance of T helper cells and regulatory T cells by enhancing the inflammatory signaling pathways to combat FA. This review will discuss the influence factors of FA, key species of gut microbiota involved in FA, types of FA, and profiles of HMOs and provide evidence for future research trends to advance HMOs as potential therapeutic aids in preventing the progress of FA.
Collapse
Affiliation(s)
- Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yuxi Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, 32004 Ourense, Spain
| | - Runfan Zhao
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Zhengxin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
Martino D, Kresoje N, Amenyogbe N, Ben-Othman R, Cai B, Lo M, Idoko O, Odumade OA, Falsafi R, Blimkie TM, An A, Shannon CP, Montante S, Dhillon BK, Diray-Arce J, Ozonoff A, Smolen KK, Brinkman RR, McEnaney K, Angelidou A, Richmond P, Tebbutt SJ, Kampmann B, Levy O, Hancock REW, Lee AHY, Kollmann TR. DNA Methylation signatures underpinning blood neutrophil to lymphocyte ratio during first week of human life. Nat Commun 2024; 15:8167. [PMID: 39289350 PMCID: PMC11408723 DOI: 10.1038/s41467-024-52283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Understanding of newborn immune ontogeny in the first week of life will enable age-appropriate strategies for safeguarding vulnerable newborns against infectious diseases. Here we conducted an observational study exploring the immunological profile of infants longitudinally throughout their first week of life. Our Expanded Program on Immunization - Human Immunology Project Consortium (EPIC-HIPC) studies the epigenetic regulation of systemic immunity using small volumes of peripheral blood samples collected from West African neonates on days of life (DOL) 0, 1, 3, and 7. Genome-wide DNA methylation and single nucleotide polymorphism markers are examined alongside matched transcriptomic and flow cytometric data. Integrative analysis reveals that a core network of transcription factors mediates dynamic shifts in neutrophil-to-lymphocyte ratios (NLR), which are underpinned by cell-type specific methylation patterns in the two cell types. Genetic variants are associated with lower NLRs at birth, and healthy newborns with lower NLRs at birth are more likely to subsequently develop sepsis. These findings provide valuable insights into the early-life determinants of immune system development.
Collapse
Affiliation(s)
- David Martino
- The Kids Research Institute Australia, Perth, WA, Australia.
- University of Western Australia, Crawley, WA, Australia.
| | - Nina Kresoje
- The Kids Research Institute Australia, Perth, WA, Australia
| | - Nelly Amenyogbe
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Bing Cai
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Mandy Lo
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Olubukola Idoko
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Reza Falsafi
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Travis M Blimkie
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Andy An
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Casey P Shannon
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | | | - Bhavjinder K Dhillon
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Asimenia Angelidou
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neonatology, Beth Israel Deaconess Medical Centre, Boston, MA, USA
| | - Peter Richmond
- The Kids Research Institute Australia, Perth, WA, Australia
- University of Western Australia, Crawley, WA, Australia
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Beate Kampmann
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
- Centre for Global Health and Institute for International Health, Charite Universitatsmedizin, Berlin, Germany
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E W Hancock
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Tobias R Kollmann
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
6
|
Qazi KR, Govindaraj D, Martí M, de Jong Y, Jensen GB, Abrahamsson T, Jenmalm MC, Sverremark-Ekström E. Impact of Extreme Prematurity, Chorioamnionitis, and Sepsis on Neonatal Monocyte Characteristics and Functions. J Innate Immun 2024; 16:470-488. [PMID: 39278208 PMCID: PMC11521501 DOI: 10.1159/000541468] [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: 04/19/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION The innate branch of the immune system is important in early life, in particular for infants born preterm. METHODS We performed a longitudinal analysis of the peripheral monocyte compartment in extremely preterm children from a randomized, placebo-controlled study of probiotic supplementation. PBMCs and fecal samples were collected at several timepoints during the first months of life. Monocyte characteristics were analyzed by flow cytometry, and LPS-stimulated PBMC culture supernatants were analyzed by Luminex or ELISA. Plasma cytokines and gut microbiota composition were analyzed by ELISA and 16S rRNA-sequencing, respectively. RESULTS The extremely preterm infants had persistent alterations in their monocyte characteristics that were further aggravated in chorioamnionitis cases. They showed a markedly reduced TLR4 expression and hampered LPS-stimulated cytokine responses 14 days after birth. Notably, at later timepoints, TLR4 expression and LPS responses no longer correlated. Sepsis during the first weeks of life strongly associated with increased pro-inflammatory, and reduced IL-10, responses also at postmenstrual week 36. Further, we report a correlation between gut microbiota features and monocyte phenotype and responses, but also that probiotic supplementation associated with distinct monocyte phenotypic characteristics, without significantly influencing their responsiveness. CONCLUSION Extremely preterm infants have monocyte characteristics and functional features that deviate from infants born full-term. Some of these differences persist until they reach an age corresponding to full-term, potentially making them more vulnerable to microbial exposures during the first months of life.
Collapse
Affiliation(s)
- Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Dhanapal Govindaraj
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Magalí Martí
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ymke de Jong
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Georg Bach Jensen
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Thomas Abrahamsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Maria C. Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
7
|
Chen CJ, Yi H, Stanley N. Conditional Similarity Triplets Enable Covariate-Informed Representations of Single-Cell Data. RESEARCH SQUARE 2024:rs.3.rs-4915088. [PMID: 39315265 PMCID: PMC11419254 DOI: 10.21203/rs.3.rs-4915088/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Single-cell technologies enable comprehensive profiling of diverse immune cell-types through the measurement of multiple genes or proteins per cell. In order to translate data from immune profiling assays into powerful diagnostics, machine learning approaches are used to compute per-sample immunological summaries, or featurizations that can be used as inputs to models for outcomes of interest. Current supervised learning approaches for computing per-sample representations are optimized based only on the outcome variable to be predicted and do not take into account clinically-relevant covariates that are likely to also be measured. Here we expand the optimization problem to also take into account such additional patient covariates to directly inform the learned per-sample representations. To do this, we introduce CytoCoSet, a set-based encoding method, which formulates a loss function with an additional triplet term penalizing samples with similar covariates from having disparate embedding results in per-sample representations. Overall, incorporating clinical covariates leads to improved prediction of clinical phenotypes.
Collapse
Affiliation(s)
- Chi-Jane Chen
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haidong Yi
- Department of Computer Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie Stanley
- Department of Computer Science and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Mithal LB, Lancki N, Ling-Hu T, Goo YA, Otero S, Rhodes NJ, Cho BK, Grobman WA, Hultquist JF, Scholtens D, Mestan KG, Seed PC. Evolution of the Umbilical Cord Blood Proteome Across Gestational Development. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.21.24309280. [PMID: 38947010 PMCID: PMC11213116 DOI: 10.1101/2024.06.21.24309280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Neonatal health is dependent on early risk stratification, diagnosis, and timely management of potentially devastating conditions, particularly in the setting of prematurity. Many of these conditions are poorly predicted in real-time by clinical data and current diagnostics. Umbilical cord blood may represent a novel source of molecular signatures that provides a window into the state of the fetus at birth. In this study, we comprehensively characterized the cord blood proteome of infants born between 24 to 42 weeks using untargeted mass spectrometry and functional enrichment analysis. We determined that the cord blood proteome at birth varies significantly across gestational development. Proteins that function in structural development and growth (e.g., extracellular matrix organization, lipid particle remodeling, and blood vessel development) are more abundant earlier in gestation. In later gestations, proteins with increased abundance are in immune response and inflammatory pathways, including complements and calcium-binding proteins. Furthermore, these data contribute to the knowledge of the physiologic state of neonates across gestational age, which is crucial to understand as we strive to best support postnatal development in preterm infants, determine mechanisms of pathology causing adverse health outcomes, and develop cord blood biomarkers to help tailor our diagnosis and therapeutics for critical neonatal conditions.
Collapse
Affiliation(s)
- Leena B. Mithal
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicola Lancki
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ted Ling-Hu
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Young Ah Goo
- Mass Spectrometry Technology Access Center at McDonnell Genome Institute (MTAC@MGI), Washington University in Saint Louis School of Medicine, MO, USA
| | - Sebastian Otero
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nathaniel J. Rhodes
- Department of Pharmacy Practice, Midwestern University, College of Pharmacy, Downers Grove, IL, USA
- Pharmacometrics Center of Excellence, Midwestern University, Downers Grove, IL, USA
- Department of Pharmacy, Northwestern Memorial Hospital, Chicago, IL, USA
| | - Byoung-Kyu Cho
- Mass Spectrometry Technology Access Center at McDonnell Genome Institute (MTAC@MGI), Washington University in Saint Louis School of Medicine, MO, USA
| | - William A. Grobman
- Department of Obstetrics and Gynecology, Ohio State University, Columbus, OH, USA
| | - Judd F. Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise Scholtens
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karen G. Mestan
- Department of Pediatrics, Division of Neonatology, University of California San Diego, CA, USA
| | - Patrick C. Seed
- Department of Pediatrics, Division of Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
9
|
Das A, Ariyakumar G, Gupta N, Kamdar S, Barugahare A, Deveson-Lucas D, Gee S, Costeloe K, Davey MS, Fleming P, Gibbons DL. Identifying immune signatures of sepsis to increase diagnostic accuracy in very preterm babies. Nat Commun 2024; 15:388. [PMID: 38195661 PMCID: PMC10776581 DOI: 10.1038/s41467-023-44387-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Bacterial infections are a major cause of mortality in preterm babies, yet our understanding of early-life disease-associated immune dysregulation remains limited. Here, we combine multi-parameter flow cytometry, single-cell RNA sequencing and plasma analysis to longitudinally profile blood from very preterm babies (<32 weeks gestation) across episodes of invasive bacterial infection (sepsis). We identify a dynamically changing blood immune signature of sepsis, including lymphopenia, reduced dendritic cell frequencies and myeloid cell HLA-DR expression, which characterizes sepsis even when the common clinical marker of inflammation, C-reactive protein, is not elevated. Furthermore, single-cell RNA sequencing identifies upregulation of amphiregulin in leukocyte populations during sepsis, which we validate as a plasma analyte that correlates with clinical signs of disease, even when C-reactive protein is normal. This study provides insights into immune pathways associated with early-life sepsis and identifies immune analytes as potential diagnostic adjuncts to standard tests to guide targeted antibiotic prescribing.
Collapse
Affiliation(s)
- A Das
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK.
| | - G Ariyakumar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - N Gupta
- Department of Neonatology, Evelina London Neonatal Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - S Kamdar
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - A Barugahare
- Bioinformatics Platform and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - D Deveson-Lucas
- Bioinformatics Platform and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - S Gee
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - K Costeloe
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - M S Davey
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - P Fleming
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Neonatology, Homerton Healthcare NHS Foundation Trust, London, UK
| | - D L Gibbons
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
10
|
Yi H, Stanley N. scLKME: A Landmark-based Approach for Generating Multi-cellular Sample Embeddings from Single-cell Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566846. [PMID: 38014189 PMCID: PMC10680595 DOI: 10.1101/2023.11.13.566846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Single-cell technologies enable high-dimensional profiling of individual cells, therefore offering profound insights into subtle variation between specialized cell-types. However, translating the multitude of nuanced cellular profiles into meaningful per-sample representations is challenging due to heterogeneous cellular composition across individual profiled samples. To compute informative per-sample representations, we developed scLKME, a novel approach that uses a landmark-based kernel mean embedding method to convert multi-sample single-cell data into compact per-sample embeddings. Treating each sample as a distribution over cells, scLKME identifies landmarks across samples and maps these distributions into a reproducing kernel Hilbert space. Overall, scLKME outperforms state-of-the-art techniques in robustness, efficiency, accuracy, and practical usefulness of sample embeddings. Its application on a CyTOF dataset profiling immune responses in preterm birth highlighted its capacity to accurately identify patient-specific variations correlating with gestational age, suggesting broad applicability to multi-sample single-cell datasets with complex experimental designs. scLKME is available as an open-sourced python package at https://github.com/CompCy-lab/scLKME.
Collapse
Affiliation(s)
- Haidong Yi
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| | - Natalie Stanley
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
- Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill NC 27599, USA
| |
Collapse
|
11
|
Ma H, Zhao W, Song T, Baijiu Z, Zhang Z. Comparative Analysis of the Pre-Parturition and Post-Parturition Genital Tract Microbiota in Plateau Bangor Sewa Sheep. Vet Sci 2023; 10:523. [PMID: 37624310 PMCID: PMC10459245 DOI: 10.3390/vetsci10080523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
(1) Background: Bangor Sewa sheep are an economically significant livestock species on the plateau. The roles of microbiota in reproduction are complex and critical for animal health. But little is known currently about the microbiome of plateau Bangor Sewa sheep. The purpose of this study was to discover the changes in the genital tract microbiota of pre- and post-partum Bangor Sewa sheep. (2) Methods: Samples from the birth canal were obtained for 16S rRNA sequencing, three days before and after delivery, respectively. (3) Results: The results showed that there was a noticeable difference in three phyla and 74 genera between the pre- and post-parturition groups in the microbiota of Bangor Sewa sheep. The changes included a decrease in the abundance of genera related to health (unclassified_Cellulomonadaceae, Cellulomonas, Fibrobacti, Flavobacterium, Eubacterium_ventriosum_group, Acetitomaculum, Aeromicrobium, Dietzia, Romboutsia, Ruminococcus, etc.) and an increased abundance of negatively related genera (Nocardioides, unclassified_Clostridia, Sphingobacteriaceae, unclassified_Ruminococcaceae, Prevotellaceae_UCG_004, Micromonospora, Streptococcus, Facklamia, Bosea, etc.) spp. (4) Conclusions: Microbes can serve as indicators of the physical state of Bangor Sewa sheep. These findings laid the foundation for deciphering the effects of microbial changes during birth on the reproductive health of plateau Bangor Sewa sheep.
Collapse
Affiliation(s)
- Hongcai Ma
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Tianzeng Song
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Zhaxi Baijiu
- Cultural Service Center of Maqian Township, Nagqu 852599, China;
| | - Zhenzhen Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| |
Collapse
|
12
|
Boeckel H, Karsten CM, Göpel W, Herting E, Rupp J, Härtel C, Hartz A. Increased Expression of Anaphylatoxin C5a-Receptor-1 in Neutrophils and Natural Killer Cells of Preterm Infants. Int J Mol Sci 2023; 24:10321. [PMID: 37373467 DOI: 10.3390/ijms241210321] [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: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Preterm infants are susceptible to infection and their defense against pathogens relies largely on innate immunity. The role of the complement system for the immunological vulnerability of preterm infants is less understood. Anaphylatoxin C5a and its receptors C5aR1 and -2 are known to be involved in sepsis pathogenesis, with C5aR1 mainly exerting pro-inflammatory effects. Our explorative study aimed to determine age-dependent changes in the expression of C5aR1 and C5aR2 in neonatal immune cell subsets. Via flow cytometry, we analyzed the expression pattern of C5a receptors on immune cells isolated from peripheral blood of preterm infants (n = 32) compared to those of their mothers (n = 25). Term infants and healthy adults served as controls. Preterm infants had a higher intracellular expression of C5aR1 on neutrophils than control individuals. We also found a higher expression of C5aR1 on NK cells, particularly on the cytotoxic CD56dim subset and the CD56- subset. Immune phenotyping of other leukocyte subpopulations revealed no gestational-age-related differences for the expression of and C5aR2. Elevated expression of C5aR1 on neutrophils and NK cells in preterm infants may contribute to the phenomenon of "immunoparalysis" caused by complement activation or to sustained hyper-inflammatory states. Further functional analyses are needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Hannah Boeckel
- Department of Pediatrics, University of Lübeck, 23538 Lübeck, Germany
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
| | - Christian M Karsten
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
- Institute for Systemic Inflammation Medicine, University of Lübeck, 23538 Lübeck, Germany
| | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck, 23538 Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck, 23538 Lübeck, Germany
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
- German Center of Infection Research, Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Jan Rupp
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
- German Center of Infection Research, Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - Christoph Härtel
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
- German Center of Infection Research, Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
- Interdisciplinary Center of Clinical Research, University of Würzburg, 97080 Würzburg, Germany
- Department of Pediatrics, University of Würzburg, 97080 Würzburg, Germany
| | - Annika Hartz
- Department of Pediatrics, University of Lübeck, 23538 Lübeck, Germany
- International Research Training Group 1911, University of Lübeck, 23538 Lübeck, Germany
- Institute for Systemic Inflammation Medicine, University of Lübeck, 23538 Lübeck, Germany
- German Center of Infection Research, Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| |
Collapse
|
13
|
Balázs G, Balajthy A, Seri I, Hegyi T, Ertl T, Szabó T, Röszer T, Papp Á, Balla J, Gáll T, Balla G. Prevention of Chronic Morbidities in Extremely Premature Newborns with LISA-nCPAP Respiratory Therapy and Adjuvant Perinatal Strategies. Antioxidants (Basel) 2023; 12:1149. [PMID: 37371878 DOI: 10.3390/antiox12061149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Less invasive surfactant administration techniques, together with nasal continuous airway pressure (LISA-nCPAP) ventilation, an emerging noninvasive ventilation (NIV) technique in neonatology, are gaining more significance, even in extremely premature newborns (ELBW), under 27 weeks of gestational age. In this review, studies on LISA-nCPAP are compiled with an emphasis on short- and long-term morbidities associated with prematurity. Several perinatal preventative and therapeutic investigations are also discussed in order to start integrated therapies as numerous organ-saving techniques in addition to lung-protective ventilations. Two thirds of immature newborns can start their lives on NIV, and one third of them never need mechanical ventilation. With adjuvant intervention, these ratios are expected to be increased, resulting in better outcomes. Optimized cardiopulmonary transition, especially physiologic cord clamping, could have an additively beneficial effect on patient outcomes gained from NIV. Organ development and angiogenesis are strictly linked not only in the immature lung and retina, but also possibly in the kidney, and optimized interventions using angiogenic growth factors could lead to better morbidity-free survival. Corticosteroids, caffeine, insulin, thyroid hormones, antioxidants, N-acetylcysteine, and, moreover, the immunomodulatory components of mother's milk are also discussed as adjuvant treatments, since immature newborns deserve more complex neonatal interventions.
Collapse
Affiliation(s)
- Gergely Balázs
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - András Balajthy
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - István Seri
- First Department of Pediatrics, School of Medicine, Semmelweis University, 1083 Budapest, Hungary
- Keck School of Medicine of USC, Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Thomas Hegyi
- Department of Pediatrics, Division of Neonatology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Tibor Ertl
- Departments of Neonatology and Obstetrics & Gynecology, University of Pécs Medical School, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Papp
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Gáll
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - György Balla
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
14
|
Ozen M, Aghaeepour N, Marić I, Wong RJ, Stevenson DK, Jantzie LL. Omics approaches: interactions at the maternal-fetal interface and origins of child health and disease. Pediatr Res 2023; 93:366-375. [PMID: 36216868 PMCID: PMC9549444 DOI: 10.1038/s41390-022-02335-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/08/2022] [Accepted: 09/18/2022] [Indexed: 11/09/2022]
Abstract
Immunoperinatology is an emerging field. Transdisciplinary efforts by physicians, physician-scientists, basic science researchers, and computational biologists have made substantial advancements by identifying unique immunologic signatures of specific diseases, discovering innovative preventative or treatment strategies, and establishing foundations for individualized neonatal intensive care of the most vulnerable neonates. In this review, we summarize the immunobiology and immunopathology of pregnancy, highlight omics approaches to study the maternal-fetal interface, and their contributions to pregnancy health. We examined the importance of transdisciplinary, multiomic (such as genomics, transcriptomics, proteomics, metabolomics, and immunomics) and machine-learning strategies in unraveling the mechanisms of adverse pregnancy, neonatal, and childhood outcomes and how they can guide the development of novel therapies to improve maternal and neonatal health. IMPACT: Discuss immunoperinatology research from the lens of omics and machine-learning approaches. Identify opportunities for omics-based approaches to delineate infection/inflammation-associated maternal, neonatal, and later life adverse outcomes (e.g., histologic chorioamnionitis [HCA]).
Collapse
Affiliation(s)
- Maide Ozen
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Nima Aghaeepour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren L Jantzie
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Al Gharaibeh FN, Kempton KM, Alder MN. Olfactomedin-4-Positive Neutrophils in Neonates: Link to Systemic Inflammation and Bronchopulmonary Dysplasia. Neonatology 2022; 120:40-48. [PMID: 36549285 PMCID: PMC10010669 DOI: 10.1159/000527902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/21/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Little is known about the interplay between neutrophil heterogeneity in neonates in health and disease states. Olfactomedin-4 (OLFM4) marks a subset of neutrophils that have been described in adults and pediatric patients but not neonates, and this subset is thought to play a role in modulating the host inflammatory response. METHODS This is a prospective cohort of neonates who were born between June 2020 and December 2021 at the University of Cincinnati Medical Center NICU. Olfactomedin-4-positive (OLFM4+) neutrophils were identified in the peripheral blood using flow cytometry. RESULTS OLFM4+ neutrophil percentage was not correlated with gestational age or developmental age. Neonates with sepsis had a higher percentage than those without the condition, 66.9% (IQR 24.3-76.9%) versus 21.5% (IQR 10.6-34.7%), respectively, p = 0.0003. At birth, a high percentage of OLFM4+ neutrophils was associated with severe chorioamnionitis at 49.1% (IQR 28.2-61.5%) compared to those without it at 13.7% (IQR 7.7-26.3%), p < 0.0001. Among neonates without sepsis, the percentages of OLFM4+ neutrophils were lower in the BPD/early death group compared to those without BPD, 11.8% (IQR 6.3-29.0%) versus 32.5% (IQR 18.5-46.1%), p = 0.003, and this retained significance in a multiple logistic regression model that included gestational age, birthweight, and race. CONCLUSION This is the first study describing OLFM4+ neutrophils in neonates and it shows that this neutrophil subpopulation is not influenced by gestational age but is elevated in inflammatory conditions such as sepsis and severe chorioamnionitis, and lower percentage at birth is associated with developing bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Faris N Al Gharaibeh
- Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kristalynn M Kempton
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Matthew N Alder
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
16
|
Huang EE, Zhang N, Ganio EA, Shen H, Li X, Ueno M, Utsunomiya T, Maruyama M, Gao Q, Su N, Yao Z, Yang F, Gaudillière B, Goodman SB. Differential dynamics of bone graft transplantation and mesenchymal stem cell therapy during bone defect healing in a murine critical size defect. J Orthop Translat 2022; 36:64-74. [PMID: 35979174 PMCID: PMC9357712 DOI: 10.1016/j.jot.2022.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 10/24/2022] Open
Abstract
Background A critical size bone defect is a clinical scenario in which bone is lost or excised due to trauma, infection, tumor, or other causes, and cannot completely heal spontaneously. The most common treatment for this condition is autologous bone grafting to the defect site. However, autologous bone graft is often insufficient in quantity or quality for transplantation to these large defects. Recently, tissue engineering methods using mesenchymal stem cells (MSCs) have been proposed as an alternative treatment. However, the underlying biological principles and optimal techniques for tissue regeneration of bone using stem cell therapy have not been completely elucidated. Methods In this study, we compare the early cellular dynamics of healing between bone graft transplantation and MSC therapy in a murine chronic femoral critical-size bone defect. We employ high-dimensional mass cytometry to provide a comprehensive view of the differences in cell composition, stem cell functionality, and immunomodulatory activity between these two treatment methods one week after transplantation. Results We reveal distinct cell compositions among tissues from bone defect sites compared with original bone graft, show active recruitment of MSCs to the bone defect sites, and demonstrate the phenotypic diversity of macrophages and T cells in each group that may affect the clinical outcome. Conclusion Our results provide critical data and future directions on the use of MSCs for treating critical size defects to regenerate bone.Translational Potential of this article: This study showed systematic comparisons of the cellular and immunomodulatory profiles among different interventions to improve the healing of the critical-size bone defect. The results provided potential strategies for designing robust therapeutic interventions for the unmet clinical need of treating critical-size bone defects.
Collapse
Affiliation(s)
- Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Edward A. Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Ni Su
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
17
|
McDavid A, Laniewski N, Grier A, Gill AL, Kessler HA, Huyck H, Carbonell E, Holden-Wiltse J, Bandyopadhyay S, Carnahan J, Dylag AM, Topham DJ, Falsey AR, Caserta MT, Pryhuber GS, Gill SR, Scheible KM. Aberrant newborn T cell and microbiota developmental trajectories predict respiratory compromise during infancy. iScience 2022; 25:104007. [PMID: 35310935 PMCID: PMC8931366 DOI: 10.1016/j.isci.2022.104007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 11/11/2022] Open
Abstract
Neonatal immune-microbiota co-development is poorly understood, yet age-appropriate recognition of - and response to - pathogens and commensal microbiota is critical to health. In this longitudinal study of 148 preterm and 119 full-term infants from birth through one year of age, we found that postmenstrual age or weeks from conception is a central factor influencing T cell and mucosal microbiota development. Numerous features of the T cell and microbiota functional development remain unexplained; however, by either age metric and are instead shaped by discrete perinatal and postnatal events. Most strikingly, we establish that prenatal antibiotics or infection disrupt the normal T cell population developmental trajectory, influencing subsequent respiratory microbial colonization and predicting respiratory morbidity. In this way, early exposures predict the postnatal immune-microbiota axis trajectory, placing infants at later risk for respiratory morbidity in early childhood.
Collapse
Affiliation(s)
- Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Nathan Laniewski
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Alex Grier
- Genomics Research Center, University of Rochester, Rochester, NY, USA
| | - Ann L. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Haeja A. Kessler
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Heidie Huyck
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Sanjukta Bandyopadhyay
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Jennifer Carnahan
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Andrew M. Dylag
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Ann R. Falsey
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Mary T. Caserta
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | | | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
18
|
Feyaerts D, Gillard J, van Cranenbroek B, Rigodanzo Marins L, Baghdady MMS, Comitini G, Lely AT, van Hamersvelt HW, van der Heijden OWH, Joosten I, van der Molen RG. Maternal, Decidual, and Neonatal Lymphocyte Composition Is Affected in Pregnant Kidney Transplant Recipients. Front Immunol 2021; 12:735564. [PMID: 34777345 PMCID: PMC8585145 DOI: 10.3389/fimmu.2021.735564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Pregnancy after renal transplantation is associated with an increased risk of complications. While a delicately balanced uterine immune system is essential for a successful pregnancy, little is known about the uterine immune environment of pregnant kidney transplant recipients. Moreover, children born to kidney transplant recipients are exposed in utero to immunosuppressive drugs, with possible consequences for neonatal outcomes. Here, we defined the effects of kidney transplantation on the immune cell composition during pregnancy with a cohort of kidney transplant recipients as well as healthy controls with uncomplicated pregnancies. Maternal immune cells from peripheral blood were collected during pregnancy as well as from decidua and cord blood obtained after delivery. Multiparameter flow cytometry was used to identify and characterize populations of cells. While systemic immune cell frequencies were altered in kidney transplant patients, immune cell dynamics over the course of pregnancy were largely similar to healthy women. In the decidua of women with a kidney transplant, we observed a decreased frequency of HLA-DR+ Treg, particularly in those treated with tacrolimus versus those that were treated with azathioprine next to tacrolimus, or with azathioprine alone. In addition, both the innate and adaptive neonatal immune system of children born to kidney transplant recipients was significantly altered compared to neonates born from uncomplicated pregnancies. Overall, our findings indicate a significant and distinct impact on the maternal systemic, uterine, and neonatal immune cell composition in pregnant kidney transplant recipients, which could have important consequences for the incidence of pregnancy complications, treatment decisions, and the offspring's health.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joshua Gillard
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Section Pediatric Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bram van Cranenbroek
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lina Rigodanzo Marins
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Gynecology and Obstetrics, Hospital de Clinicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Mariam M S Baghdady
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gaia Comitini
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - A Titia Lely
- Department of Obstetrics and Gynecology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | - Irma Joosten
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Renate G van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|