1
|
Shi Y, Li K, Ding R, Li X, Cheng Z, Liu J, Liu S, Zhu H, Sun H. Untargeted metabolomics and machine learning unveil the exposome and metabolism linked with the risk of early pregnancy loss. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137362. [PMID: 39892135 DOI: 10.1016/j.jhazmat.2025.137362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Early pregnancy loss (EPL) may result from exposure to emerging contaminants (ECs), although the underlying mechanisms remain poorly understood. This case-control study measured over 2000 serum features, including 37 ECs, 6 biochemicals, and 2057 endogenous metabolites, in serum samples collected from 48 EPL patients and healthy pregnant women. The median total concentration of targeted EC in the EPL group (65.9 ng/mL) was significantly higher than in controls (43.0 ng/mL; p < 0.05). Four machine learning algorithms were employed to identify key molecular features and develop EPL risk prediction models. A random forest model based on chemical data achieved a predictive accuracy of 95 %, suggesting a potential association between EPL and chemical exposure, with phthalic acid esters identified as significant contributors. Ninety-five potential metabolite biomarkers were selected, which were predominantly enriched in pathways related to spermidine and spermine biosynthesis, ubiquinone biosynthesis, and pantothenate and coenzyme A biosynthesis. C17-sphinganine was identified as a leading biomarker with an area under the curve of 0.93. Furthermore, exposure to bis(2-ethylhexyl)phthalate was linked to an increased risk of EPL by disrupting lipid metabolism. These findings indicate that combining untargeted metabolomics with machine learning approaches offers novel insights into the mechanisms of EPL related to EC exposure.
Collapse
Affiliation(s)
- Yixuan Shi
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Keyi Li
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Ran Ding
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xiaoying Li
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian 116026, China.
| | - Zhipeng Cheng
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Jialan Liu
- Department of Obstetrics and Gynecology, Tianjin Jinnan Hospital, Tianjin 300350, China
| | - Shaoxia Liu
- Department of Obstetrics and Gynecology, Tianjin Jinnan Hospital, Tianjin 300350, China
| | - Hongkai Zhu
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| | - Hongwen Sun
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
2
|
Bosco A, Arru F, Abis A, Fanos V, Dessì A. Application of Multiomics in Perinatology: A Metabolomics Integration-Focused Review. Int J Mol Sci 2025; 26:4164. [PMID: 40362403 PMCID: PMC12071719 DOI: 10.3390/ijms26094164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/13/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Precision medicine stems from a new approach to the prevention, diagnosis and treatment of patients, due to the shift in focus away from pathology and towards the uniqueness of the individual, personalising the diagnostic-therapeutic pathway. This paradigm shift has been made possible by the emergence of new high-throughput technologies capable of generating large amounts of data on multiple levels of a biological system, identifying pathology-related genes, transcripts, proteins and metabolites. Metabolomics plays a primary role in this context, providing, through non-invasive sampling, a very close image of the phenotype of the organism being studied by detecting metabolites, end products downstream of gene transcription, present in cells, tissues, organs and biological fluids. The enormous amount of data that these modern technologies make available, together with the need to elucidate the complex interplay of the various biological levels by combining data from distinct omics, has led to the need to employ advanced informatics techniques, among which artificial intelligence has recently emerged. These innovations are of great interest in the field of perinatology, representing an attempt to optimise the diagnostic timeline for the most critical newborns. In addition, they may contribute to the improvement of prevention strategies available to date. All these contributions prove to be crucial at very vulnerable life stages, allowing crucial intervention opportunities. In this review, we have analysed studies that have integrated metabolomics with at least one other omics in the perinatal field, attempting to highlight the usefulness of multiomics integration and the different methods employed.
Collapse
Affiliation(s)
| | | | | | | | - Angelica Dessì
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, AOU Cagliari, 09124 Cagliari, Italy; (A.B.); (F.A.); (A.A.); (V.F.)
| |
Collapse
|
3
|
Qu J, Mao W, Chen M, Jin H. Prediction of p-phenylenediamine antioxidant concentrations in human urine using machine learning models. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137184. [PMID: 39813931 DOI: 10.1016/j.jhazmat.2025.137184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
p-phenylenediamine antioxidants (PPDs) are extensively used in rubber manufacturing for their potent antioxidative properties, but PPDs and 2-anilino-5-[(4-methylpentan-2yl)amino]cyclohexa-2,5-diene-1,4-dione (6PPDQ) pose potential environmental and health risks. Existing biomonitoring methods for assessing human exposure to PPDs are labor-intensive, costly, and provide limited data. Thus, there is a critical need to develop predictive models for evaluating PPDs and 6PPDQ exposure levels to facilitate health risk assessments. In this study, machine learning (ML) models were developed to predict the concentration of three PPDs and 6PPDQ in human urine samples. A total of 759 participants from three cities in Zhejiang Province, China, provided urine samples, which were analyzed for PPDs and 6PPDQ concentrations using liquid chromatography-tandem mass spectrometry. Eight ML models were employed to predict PPDs and 6PPDQ concentrations based on demographic and environmental exposure factors such as age, gender, body mass index (BMI), and occupation. N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD) was the most frequently detected PPD (mean 3.03 ng/mL, range < LOD-18.65 ng/mL), followed by 6PPDQ (mean 2.76 ng/mL, range < LOD-20.85 ng/mL) and N-phenyl-N'-cyclohexyl-p-phenylenediamine (mean 2.04 ng/mL, range < LOD-10.22 ng/mL). Random forest model demonstrated the highest accuracy in predicting PPDs and 6PPDQ concentrations in human urine among the ML models evaluated. Through the application of these models, age, BMI, and occupation emerged as significant predictors of urinary PPDs and 6PPDQ concentrations. This research significantly contributes by using ML models to enhance exposure assessment accuracy and efficiency, providing a novel framework for future studies on environmental health risks related to PPDs and 6PPDQ.
Collapse
Affiliation(s)
- Jianli Qu
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, PR China
| | - Weili Mao
- Department of Pharmacy, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, PR China
| | - Mei Chen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, PR China.
| |
Collapse
|
4
|
Diop M, Davidson BR, Fragiadakis GK, Sirota M, Gaudillière B, Combes AJ. Single-cell omics technologies - Fundamentals on how to create single-cell looking glasses for reproductive health. Am J Obstet Gynecol 2025; 232:S1-S20. [PMID: 40253074 DOI: 10.1016/j.ajog.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Over the last decade, in line with the goals of precision medicine to offer individualized patient care, various single-cell technologies measuring gene and proteomic expression in various tissues have rapidly advanced to study health and disease at the single cell level. Precisely understanding cell composition, position within tissues, signaling pathways, and communication can reveal insights into disease mechanisms and systemic changes during development, pregnancy, and gynecologic disorders across the lifespan. Single-cell technologies dissect the complex cellular compositions of reproductive tract tissues, providing insights into mechanisms behind reproductive tract dysfunction which impact wellness and quality of life. These technologies aim to understand basic tissue and organ functions and, clinically, to develop novel diagnostics, early disease biomarkers, and cell-targeted therapies for currently suboptimally-treated disorders. Increasingly, they are applied to pregnancy and pregnancy disorders, gynecologic malignancies, and uterine and ovarian physiology and aging, which are discussed in more detail in manuscripts in this special issue of AJOG. Here, we review recent applications of single-cell technologies to the study of gynecologic disorders and systemic biological adaptations during fetal development, pregnancy, and across a woman's lifespan. We discuss sequencing- and proteomic-based single-cell methods, as well as spatial transcriptomics and high-dimensional proteomic imaging, describing each technology's mechanism, workflow, quality control, and highlighting specific benefits, drawbacks, and utility in the context of reproductive medicine. We consider analytical methods for the high-dimensional single-cell data generated, highlighting statistical constraints and recent computational techniques for downstream clinical translation. Overall, current and evolving single-cell "looking glasses", or perspectives, have the potential to transform fundamental understanding of women's health and reproductive disorders and alter the trajectory of clinical practice and patient outcomes in the future.
Collapse
Affiliation(s)
- Maïgane Diop
- Program in Immunology, Stanford University School of Medicine, Stanford, CA; Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA
| | | | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA.
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA; Department of Pediatrics, University of California, San Francisco, CA.
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA.
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, CA; Department of Pathology, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA.
| |
Collapse
|
5
|
Korosec CS, Conway JM, Matveev VA, Ostrowski M, Heffernan JM, Ghaemi MS. Machine Learning Reveals Distinct Immunogenic Signatures of Th1 Imprinting in ART-Treated Individuals with HIV Following Repeated SARS-CoV-2 Vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643769. [PMID: 40166325 PMCID: PMC11956973 DOI: 10.1101/2025.03.18.643769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The human immune system is intrinsically variable and remarkably diverse across a population. The immune response to antigens is driven by a complex interplay of time-dependent interdependencies across components of the immune system. After repeated vaccination, the humoral and cellular arms of the immune response display highly heterogeneous dynamics, further complicating the attribution of a phenotypic outcome to specific immune system components. We employ a random forest (RF) approach to classify informative differences in immunogenicity between older people living with HIV (PLWH) on ART and an age-matched control group who received up to five SARS-CoV-2 vaccinations over 104 weeks. RFs identify immunological variables of importance, interpreted as evidence for Th1 imprinting, and suggest novel distinguishing immune features, such as saliva-based antibody screening, as promising diagnostic features towards classifying responses (whereas serum IgG is not). Additionally, we implement supervised and unsupervised Machine Learning methods to produce physiologically accurate synthetic datasets that conform to the statistical distribution of the original immunological data, thus enabling further data-driven hypothesis testing and model validation. Our results highlight the effectiveness of RFs in utilizing informative immune feature interdependencies for classification tasks and suggests broad impacts of ML applications for personalized vaccination strategies among high-risk populations.
Collapse
|
6
|
Dreisbach C, Barcelona V, Turchioe MR, Bernstein S, Erickson E. Application of Predictive Analytics in Pregnancy, Birth, and Postpartum Nursing Care. MCN Am J Matern Child Nurs 2025; 50:66-77. [PMID: 39724545 DOI: 10.1097/nmc.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
ABSTRACT Predictive analytics has emerged as a promising approach for improving reproductive health care and patient outcomes. During pregnancy and birth, the ability to accurately predict risks and complications could enable earlier interventions and reduce adverse events. However, there are challenges and ethical considerations for implementing predictive models in perinatal care settings. We introduce major concepts in predictive analytics and describe application of predictive modeling to perinatal care topics such as fertility, preeclampsia, labor onset, vaginal birth after cesarean, uterine rupture, induction outcomes, postpartum hemorrhage, and postpartum mood disorders. Although some predictive models have achieved adequate accuracy (AUC 0.7-0.9), most require additional external validation across diverse populations and practice settings. Bias, particularly racial bias, remains a key limitation of current models. Nurses and advanced practice nurses, including nurse practitioners certified registered nurse anesthetists, and nurse-midwives, play a vital role in ensuring high-quality data collection and communicating predictive model outputs to clinicians and users of the health care system. Addressing the ethical challenges and limitations of predictive analytics is imperative to equitably translate these tools to support patient-centered perinatal care.
Collapse
|
7
|
Feyaerts D, Diop M, Galaz J, Einhaus JF, Arck PC, Diemert A, Winn VD, Parast M, Gyamfi-Bannerman C, Prins JR, Gomez-Lopez N, Stelzer IA. The single-cell immune profile throughout gestation and its potential value for identifying women at risk for spontaneous preterm birth. Eur J Obstet Gynecol Reprod Biol X 2025; 25:100371. [PMID: 40052005 PMCID: PMC11883378 DOI: 10.1016/j.eurox.2025.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/23/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Precisely timed immune adaptations, observed in the maternal circulation, underpin the notion of an immune clock of human pregnancy that supports its successful progression and completion at delivery. This immune clock is divided into three immunological phases, with the first phase starting at the time of conception and implantation, shifting into the second phase that supports homeostasis and tolerance throughout pregnancy, and culminating in the last phase of labor and parturition. Disruptions of this immune clock are reported in pregnancy complications such as spontaneous preterm birth. However, our understanding of the immune clock preceding spontaneous preterm birth remains scattered. In this review, we describe the chronology of maternal immune cell adaptations during healthy pregnancies and highlight its disruption in spontaneous preterm birth. With a focus on single-cell cytometric, proteomic and transcriptomic approaches, we review recent studies of term and spontaneous preterm pregnancies and discuss the need for future prospective studies aimed at tracking pregnancies longitudinally on a multi-omic scale. Such studies will be critical in determining whether spontaneous preterm pregnancies progress at an accelerated pace or follow a preterm-intrinsic pattern when compared to those delivered at term.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Maïgane Diop
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Galaz
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jakob F. Einhaus
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Petra C. Arck
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mana Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Cynthia Gyamfi-Bannerman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jelmer R. Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nardhy Gomez-Lopez
- Departments of Obstetrics and Gynecology & Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Ina A. Stelzer
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
8
|
Zhu B, Bai Y, Yeo YY, Lu X, Rovira-Clavé X, Chen H, Yeung J, Nkosi D, Glickman J, Delgado-Gonzalez A, Gerber GK, Angelo M, Shalek AK, Nolan GP, Jiang S. A multi-omics spatial framework for host-microbiome dissection within the intestinal tissue microenvironment. Nat Commun 2025; 16:1230. [PMID: 39890778 PMCID: PMC11785740 DOI: 10.1038/s41467-025-56237-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025] Open
Abstract
The intricate interactions between the host immune system and its microbiome constituents undergo dynamic shifts in response to perturbations to the intestinal tissue environment. Our ability to study these events on the systems level is significantly limited by in situ approaches capable of generating simultaneous insights from both host and microbial communities. Here, we introduce Microbiome Cartography (MicroCart), a framework for simultaneous in situ probing of host and microbiome across multiple spatial modalities. We demonstrate MicroCart by investigating gut host and microbiome changes in a murine colitis model, using spatial proteomics, transcriptomics, and glycomics. Our findings reveal a global but systematic transformation in tissue immune responses, encompassing tissue-level remodeling in response to host immune and epithelial cell state perturbations, bacterial population shifts, localized inflammatory responses, and metabolic process alterations during colitis. MicroCart enables a deep investigation of the intricate interplay between the host tissue and its microbiome with spatial multi-omics.
Collapse
Affiliation(s)
- Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Yunhao Bai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xiaowei Lu
- Mass Spectrometry Core Facility, Stanford University, Stanford, CA, USA
| | - Xavier Rovira-Clavé
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Biological and Medical Informatics Program, UCSF, San Francisco, CA, USA
| | - Jason Yeung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dingani Nkosi
- Department of Pathology, Massachusetts General Brigham, Boston, MA, USA
| | - Jonathan Glickman
- Department of Pathology, Massachusetts General Brigham, Boston, MA, USA
| | | | - Georg K Gerber
- Division of Computational Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Health Sciences and Technology, Harvard University and MIT, Cambridge, MA, USA
| | - Mike Angelo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Sizun Jiang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Division of Computational Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Mataraso SJ, Espinosa CA, Seong D, Reincke SM, Berson E, Reiss JD, Kim Y, Ghanem M, Shu CH, James T, Tan Y, Shome S, Stelzer IA, Feyaerts D, Wong RJ, Shaw GM, Angst MS, Gaudilliere B, Stevenson DK, Aghaeepour N. A machine learning approach to leveraging electronic health records for enhanced omics analysis. NAT MACH INTELL 2025; 7:293-306. [PMID: 40008295 PMCID: PMC11847705 DOI: 10.1038/s42256-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025]
Abstract
Omics studies produce a large number of measurements, enabling the development, validation and interpretation of systems-level biological models. Large cohorts are required to power these complex models; yet, the cohort size remains limited due to clinical and budgetary constraints. We introduce clinical and omics multimodal analysis enhanced with transfer learning (COMET), a machine learning framework that incorporates large, observational electronic health record databases and transfer learning to improve the analysis of small datasets from omics studies. By pretraining on electronic health record data and adaptively blending both early and late fusion strategies, COMET overcomes the limitations of existing multimodal machine learning methods. Using two independent datasets, we showed that COMET improved the predictive modelling performance and biological discovery compared with the analysis of omics data with traditional methods. By incorporating electronic health record data into omics analyses, COMET enables more precise patient classifications, beyond the simplistic binary reduction to cases and controls. This framework can be broadly applied to the analysis of multimodal omics studies and reveals more powerful biological insights from limited cohort sizes.
Collapse
Affiliation(s)
- Samson J. Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
| | - Camilo A. Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA USA
| | - David Seong
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA USA
| | - S. Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Jonathan D. Reiss
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
| | - Yeasul Kim
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
| | - Marc Ghanem
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Tomin James
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Yuqi Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA USA
| | - Sayane Shome
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, University of California San Diego, La Jolla, CA USA
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
10
|
Wu T, Zhao L, Ren M, He S, Zhang L, Fang M, Wang B. Small-Sample Learning for Next-Generation Human Health Risk Assessment: Harnessing AI, Exposome Data, and Systems Biology. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:5-10. [PMID: 39745662 DOI: 10.1021/acs.est.4c11832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Affiliation(s)
- Tianxiang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Lu Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Mengyuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
| | - Song He
- Department of Bioinformatics, Academy of Military Medical Sciences, Beijing 100850, China
| | - Le Zhang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Bin Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of PR China, Beijing 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
- Laboratory for Earth Surface Processes, College of Urban and Environmental Science, Peking University, Beijing 100871, China
| |
Collapse
|
11
|
Basavaraj C, Grant AD, Aras SG, Erickson EN. Deep learning model using continuous skin temperature data predicts labor onset. BMC Pregnancy Childbirth 2024; 24:777. [PMID: 39587525 PMCID: PMC11587739 DOI: 10.1186/s12884-024-06862-9] [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: 08/03/2024] [Accepted: 09/25/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Changes in body temperature anticipate labor onset in numerous mammals, yet this concept has not been explored in humans. We investigated if continuous body temperature exhibits similar changes in women and whether these changes may be linked to hormonal status. Finally, we developed a deep learning model using temperature patterning to provide a daily forecast of time to labor onset. METHODS We evaluated patterns in continuous skin temperature data in 91 (n = 54 spontaneous labors) pregnant women using a wearable smart ring. In a subset of 28 pregnancies, we examined daily steroid hormone samples leading up to labor to analyze relationships among hormones and body temperature trajectory. Finally, we applied an autoencoder long short-term memory (AE-LSTM) deep learning model to provide a novel daily estimation of days until labor onset. RESULTS Features of temperature change leading up to labor were associated with urinary hormones and labor type. Spontaneous labors exhibited greater estriol to α-pregnanediol ratio, as well as lower body temperature and more stable circadian rhythms compared to pregnancies that did not undergo spontaneous labor. Skin temperature data from 54 pregnancies that underwent spontaneous labor between 34 and 42 weeks of gestation were included in training the AE-LSTM model, and an additional 37 pregnancies that underwent artificial induction of labor or Cesarean without labor were used for further testing. The input to the pipeline was 5-min skin temperature data from a gestational age of 240 days until the day of labor onset. During cross-validation AE-LSTM average error (true - predicted) dropped below 2 days at 8 days before labor, independent of gestational age. Labor onset windows were calculated from the AE-LSTM output using a probabilistic distribution of model error. For these windows AE-LSTM correctly predicted labor start for 79% of the spontaneous labors within a 4.6-day window at 7 days before true labor, and 7.4-day window at 10 days before true labor. CONCLUSION Continuous skin temperature reflects progression toward labor and hormonal change during pregnancy. Deep learning using continuous temperature may provide clinically valuable tools for pregnancy care.
Collapse
Affiliation(s)
- Chinmai Basavaraj
- Department of Computer Science, The University of Arizona, Tucson, AZ, USA
| | | | - Shravan G Aras
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
12
|
Stevenson DK, Chang AL, Wong RJ, Reiss JD, Gaudillière B, Sylvester KG, Ling XB, Angst MS, Shaw GM, Katz M, Aghaeepour N, Marić I. Towards a new taxonomy of preterm birth. J Perinatol 2024:10.1038/s41372-024-02183-z. [PMID: 39567650 DOI: 10.1038/s41372-024-02183-z] [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: 10/19/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Disease categories traditionally reflect a historical clustering of clinical phenotypes based on biologic and nonbiologic features. Multiomics approaches have striven to identify signatures to develop individualized categorizations through tests and/or therapies for 'personalized' medicine. Precision health classifies clinical syndromes into endotype clusters based on novel technological advancements, which can reveal insights into the etiologies of phenotypical syndromes. A new taxonomy of preterm birth should be considered in this context, as not all preterm infants of similar gestational ages are the same because most have different biologic vulnerabilities and hence different health trajectories. Even the choice of interventions may affect observed clinical conditions. Thus, a new taxonomy of prematurity would help to advance the field of neonatology, but also obstetrics and perinatology by adopting anticipatory and more targeted approaches to the care of preterm infants with the intent of preventing and treating some of the most common newborn pathologic conditions.
Collapse
Affiliation(s)
- David K Stevenson
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Alan L Chang
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan D Reiss
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuefeng B Ling
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Gary M Shaw
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Katz
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
13
|
Murphy KR, Farrell JS, Bendig J, Mitra A, Luff C, Stelzer IA, Yamaguchi H, Angelakos CC, Choi M, Bian W, DiIanni T, Pujol EM, Matosevich N, Airan R, Gaudillière B, Konofagou EE, Butts-Pauly K, Soltesz I, de Lecea L. Optimized ultrasound neuromodulation for non-invasive control of behavior and physiology. Neuron 2024; 112:3252-3266.e5. [PMID: 39079529 PMCID: PMC11709124 DOI: 10.1016/j.neuron.2024.07.002] [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: 11/27/2023] [Revised: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 08/09/2024]
Abstract
Focused ultrasound can non-invasively modulate neural activity, but whether effective stimulation parameters generalize across brain regions and cell types remains unknown. We used focused ultrasound coupled with fiber photometry to identify optimal neuromodulation parameters for four different arousal centers of the brain in an effort to yield overt changes in behavior. Applying coordinate descent, we found that optimal parameters for excitation or inhibition are highly distinct, the effects of which are generally conserved across brain regions and cell types. Optimized stimulations induced clear, target-specific behavioral effects, whereas non-optimized protocols of equivalent energy resulted in substantially less or no change in behavior. These outcomes were independent of auditory confounds and, contrary to expectation, accompanied by a cyclooxygenase-dependent and prolonged reduction in local blood flow and temperature with brain-region-specific scaling. These findings demonstrate that carefully tuned and targeted ultrasound can exhibit powerful effects on complex behavior and physiology.
Collapse
Affiliation(s)
- Keith R Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Jonas Bendig
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anish Mitra
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Charlotte Luff
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Ina A Stelzer
- Department of Anesthesia, Stanford University, Stanford, CA, USA
| | - Hiroshi Yamaguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Department of Neuroscience, Nagoya University, Nagoya, Japan
| | | | - Mihyun Choi
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Wenjie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Tommaso DiIanni
- Department of Radiology, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Esther Martinez Pujol
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Noa Matosevich
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Raag Airan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kim Butts-Pauly
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
14
|
Hédou J, Marić I, Bellan G, Einhaus J, Gaudillière DK, Ladant FX, Verdonk F, Stelzer IA, Feyaerts D, Tsai AS, Ganio EA, Sabayev M, Gillard J, Amar J, Cambriel A, Oskotsky TT, Roldan A, Golob JL, Sirota M, Bonham TA, Sato M, Diop M, Durand X, Angst MS, Stevenson DK, Aghaeepour N, Montanari A, Gaudillière B. Discovery of sparse, reliable omic biomarkers with Stabl. Nat Biotechnol 2024; 42:1581-1593. [PMID: 38168992 PMCID: PMC11217152 DOI: 10.1038/s41587-023-02033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024]
Abstract
Adoption of high-content omic technologies in clinical studies, coupled with computational methods, has yielded an abundance of candidate biomarkers. However, translating such findings into bona fide clinical biomarkers remains challenging. To facilitate this process, we introduce Stabl, a general machine learning method that identifies a sparse, reliable set of biomarkers by integrating noise injection and a data-driven signal-to-noise threshold into multivariable predictive modeling. Evaluation of Stabl on synthetic datasets and five independent clinical studies demonstrates improved biomarker sparsity and reliability compared to commonly used sparsity-promoting regularization methods while maintaining predictive performance; it distills datasets containing 1,400-35,000 features down to 4-34 candidate biomarkers. Stabl extends to multi-omic integration tasks, enabling biological interpretation of complex predictive models, as it hones in on a shortlist of proteomic, metabolomic and cytometric events predicting labor onset, microbial biomarkers of pre-term birth and a pre-operative immune signature of post-surgical infections. Stabl is available at https://github.com/gregbellan/Stabl .
Collapse
Affiliation(s)
- Julien Hédou
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Grégoire Bellan
- Télécom Paris, Institut Polytechnique de Paris, Paris, France
| | - Jakob Einhaus
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Dyani K Gaudillière
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | | | - Franck Verdonk
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and Intensive Care, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Maximilian Sabayev
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Joshua Gillard
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jonas Amar
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Amelie Cambriel
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Alennie Roldan
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan L Golob
- Department of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas A Bonham
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Masaki Sato
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Maïgane Diop
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Xavier Durand
- École Polytechnique, Institut Polytechnique de Paris, Paris, France
| | - Martin S Angst
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Andrea Montanari
- Department of Statistics, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA.
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Seong D, Mataraso S, Espinosa C, Berson E, Reincke SM, Xue L, Kashiwagi C, Kim Y, Shu CH, Chung P, Ghanem M, Xie F, Wong RJ, Angst MS, Gaudilliere B, Shaw GM, Stevenson DK, Aghaeepour N. Generating pregnant patient biological profiles by deconvoluting clinical records with electronic health record foundation models. Brief Bioinform 2024; 25:bbae574. [PMID: 39545787 PMCID: PMC11565587 DOI: 10.1093/bib/bbae574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Translational biology posits a strong bi-directional link between clinical phenotypes and a patient's biological profile. By leveraging this bi-directional link, we can efficiently deconvolute pre-existing clinical information into biological profiles. However, traditional computational tools are limited in their ability to resolve this link because of the relatively small sizes of paired clinical-biological datasets for training and the high dimensionality/sparsity of tabular clinical data. Here, we use state-of-the-art foundation models (FMs) for electronic health record (EHR) data to generate proteomics profiles of pregnant patients, thereby deconvoluting pre-existing clinical information into biological profiles without the cost and effort of running large-scale traditional omics studies. We show that FM-derived representations of a patient's EHR data coupled with a fully connected neural network prediction head can generate 206 blood protein expression levels. Interestingly, these proteins were enriched for developmental pathways, while proteins not able to be generated from EHR data were enriched for metabolic pathways. Finally, we show a proteomic signature of gestational diabetes that includes proteins with established and novel links to gestational diabetes. These results showcase the power of FM-derived EHR representations in efficiently generating biological states of pregnant patients. This capability can revolutionize disease understanding and therapeutic development, offering a cost-effective, time-efficient, and less invasive alternative to traditional methods of generating proteomics.
Collapse
Affiliation(s)
- David Seong
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Medical Scientist Training Program, Stanford University School of Medicine, 1265 Welch Road, Stanford CA, 94305, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Camilo Espinosa
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - S Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Lei Xue
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Chloe Kashiwagi
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Yeasul Kim
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Philip Chung
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Marc Ghanem
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Feng Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Nima Aghaeepour
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| |
Collapse
|
16
|
Bæk O, Schaltz-Buchholzer F, Campbell A, Amenyogbe N, Campbell J, Aaby P, Benn CS, Kollmann TR. The mark of success: The role of vaccine-induced skin scar formation for BCG and smallpox vaccine-associated clinical benefits. Semin Immunopathol 2024; 46:13. [PMID: 39186134 PMCID: PMC11347488 DOI: 10.1007/s00281-024-01022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
Skin scar formation following Bacille Calmette-Guérin (BCG) or smallpox (Vaccinia) vaccination is an established marker of successful vaccination and 'vaccine take'. Potent pathogen-specific (tuberculosis; smallpox) and pathogen-agnostic (protection from diseases unrelated to the intentionally targeted pathogen) effects of BCG and smallpox vaccines hold significant translational potential. Yet despite their use for centuries, how scar formation occurs and how local skin-based events relate to systemic effects that allow these two vaccines to deliver powerful health promoting effects has not yet been determined. We review here what is known about the events occurring in the skin and place this knowledge in the context of the overall impact of these two vaccines on human health with a particular focus on maternal-child health.
Collapse
Affiliation(s)
- Ole Bæk
- University of Copenhagen, Copenhagen, Denmark
| | | | | | - Nelly Amenyogbe
- Telethon Kids Institute, Perth, Australia
- Dalhousie University, 5980 University Ave #5850, 4th floor Goldbloom Pavilion, Halifax, NS, B3K 6R8, Canada
- Bandim Health Project, Bissau, Guinea-Bissau
| | | | - Peter Aaby
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Christine Stabell Benn
- University of Southern Denmark, Copenhagen, Denmark
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Tobias R Kollmann
- Telethon Kids Institute, Perth, Australia.
- Dalhousie University, 5980 University Ave #5850, 4th floor Goldbloom Pavilion, Halifax, NS, B3K 6R8, Canada.
- Bandim Health Project, Bissau, Guinea-Bissau.
| |
Collapse
|
17
|
Zhang P, Jia Y, Song H, Fan Y, Lv Y, Geng H, Zhao Y, Cui H, Chen X. Novel biomarkers for prediction of atonic postpartum hemorrhage among 'low-risk' women in labor. Front Immunol 2024; 15:1416990. [PMID: 39055706 PMCID: PMC11269088 DOI: 10.3389/fimmu.2024.1416990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background Postpartum hemorrhage (PPH) is the primary cause of maternal mortality globally, with uterine atony being the predominant contributing factor. However, accurate prediction of PPH in the general population remains challenging due to a lack of reliable biomarkers. Methods Using retrospective cohort data, we quantified 48 cytokines in plasma samples from 40 women diagnosed with PPH caused by uterine atony. We also analyzed previously reported hemogram and coagulation parameters related to inflammatory response. The least absolute shrinkage and selection operator (LASSO) and logistic regression were applied to develop predictive models. Established models were further evaluated and temporally validated in a prospective cohort. Results Fourteen factors showed significant differences between the two groups, among which IL2Rα, IL9, MIP1β, TNFβ, CTACK, prenatal Hb, Lymph%, PLR, and LnSII were selected by LASSO to construct predictive model A. Further, by logistic regression, model B was constructed using prenatal Hb, PLR, IL2Rα, and IL9. The area under the curve (AUC) values of model A in the training set, internal validation set, and temporal validation set were 0.846 (0.757-0.934), 0.846 (0.749-0.930), and 0.875 (0.789-0.961), respectively. And the corresponding AUC values for model B were 0.805 (0.709-0.901), 0.805 (0.701-0.894), and 0.901 (0.824-0.979). Decision curve analysis results showed that both nomograms had a high net benefit for predicting atonic PPH. Conclusion We identified novel biomarkers and developed predictive models for atonic PPH in women undergoing "low-risk" vaginal delivery, providing immunological insights for further exploration of the mechanism underlying atonic PPH.
Collapse
Affiliation(s)
- Pei Zhang
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Yanju Jia
- School of Medicine, Nankai University, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Hui Song
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yifan Fan
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yan Lv
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Hao Geng
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Hongyan Cui
- School of Medicine, Nankai University, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Xu Chen
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
- Department of Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| |
Collapse
|
18
|
Stevenson DK, Gotlib IH, Buthmann JL, Marié I, Aghaeepour N, Gaudilliere B, Angst MS, Darmstadt GL, Druzin ML, Wong RJ, Shaw GM, Katz M. Stress and Its Consequences-Biological Strain. Am J Perinatol 2024; 41:1282-1284. [PMID: 35292943 DOI: 10.1055/a-1798-1602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Understanding the role of stress in pregnancy and its consequences is important, particularly given documented associations between maternal stress and preterm birth and other pathological outcomes. Physical and psychological stressors can elicit the same biological responses, known as biological strain. Chronic stressors, like poverty and racism (race-based discriminatory treatment), may create a legacy or trajectory of biological strain that no amount of coping can relieve in the absence of larger-scale socio-behavioral or societal changes. An integrative approach that takes into consideration simultaneously social and biological determinants of stress may provide the best insights into the risk of preterm birth. The most successful computational approaches and the most predictive machine-learning models are likely to be those that combine information about the stressors and the biological strain (for example, as measured by different omics) experienced during pregnancy.
Collapse
Affiliation(s)
- David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Ian H Gotlib
- Department of Psychology, Stanford University School of Humanities and Science, Stanford, California
| | - Jessica L Buthmann
- Department of Psychology, Stanford University School of Humanities and Science, Stanford, California
| | - Ivana Marié
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Gary L Darmstadt
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Maurice L Druzin
- Department of Obstetrics and Gynecology-Maternal-Fetal Medicine, Stanford University School of Medicine, Stanford, California
| | - Ronald J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Gary M Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Michael Katz
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
19
|
Shaffer Z, Romero R, Tarca AL, Galaz J, Arenas-Hernandez M, Gudicha DW, Chaiworapongsa T, Jung E, Suksai M, Theis KR, Gomez-Lopez N. The vaginal immunoproteome for the prediction of spontaneous preterm birth: A retrospective longitudinal study. eLife 2024; 13:e90943. [PMID: 38913421 PMCID: PMC11196114 DOI: 10.7554/elife.90943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Background Preterm birth is the leading cause of neonatal morbidity and mortality worldwide. Most cases of preterm birth occur spontaneously and result from preterm labor with intact (spontaneous preterm labor [sPTL]) or ruptured (preterm prelabor rupture of membranes [PPROM]) membranes. The prediction of spontaneous preterm birth (sPTB) remains underpowered due to its syndromic nature and the dearth of independent analyses of the vaginal host immune response. Thus, we conducted the largest longitudinal investigation targeting vaginal immune mediators, referred to herein as the immunoproteome, in a population at high risk for sPTB. Methods Vaginal swabs were collected across gestation from pregnant women who ultimately underwent term birth, sPTL, or PPROM. Cytokines, chemokines, growth factors, and antimicrobial peptides in the samples were quantified via specific and sensitive immunoassays. Predictive models were constructed from immune mediator concentrations. Results Throughout uncomplicated gestation, the vaginal immunoproteome harbors a cytokine network with a homeostatic profile. Yet, the vaginal immunoproteome is skewed toward a pro-inflammatory state in pregnant women who ultimately experience sPTL and PPROM. Such an inflammatory profile includes increased monocyte chemoattractants, cytokines indicative of macrophage and T-cell activation, and reduced antimicrobial proteins/peptides. The vaginal immunoproteome has improved predictive value over maternal characteristics alone for identifying women at risk for early (<34 weeks) sPTB. Conclusions The vaginal immunoproteome undergoes homeostatic changes throughout gestation and deviations from this shift are associated with sPTB. Furthermore, the vaginal immunoproteome can be leveraged as a potential biomarker for early sPTB, a subset of sPTB associated with extremely adverse neonatal outcomes. Funding This research was conducted by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS) under contract HHSN275201300006C. ALT, KRT, and NGL were supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health.
Collapse
Affiliation(s)
- Zachary Shaffer
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, University of MichiganAnn ArborUnited States
- Department of Epidemiology and Biostatistics, Michigan State UniversityEast LansingUnited States
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Computer Science, Wayne State University College of EngineeringDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de ChileSantiagoChile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Dereje W Gudicha
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Kevin R Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS)BethesdaUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| |
Collapse
|
20
|
Stevenson DK, Winn VD, Shaw GM, England SK, Wong RJ. Solving the Puzzle of Preterm Birth. Clin Perinatol 2024; 51:291-300. [PMID: 38705641 DOI: 10.1016/j.clp.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Solving the puzzle of preterm birth has been challenging and will require novel integrative solutions as preterm birth likely arises from many etiologies. It has been demonstrated that many sociodemographic and psychological determinants of preterm birth relate to its complex biology. It is this understanding that has enabled the development of a novel preventative strategy, which integrates the omics profile (genome, epigenome, transcriptome, proteome, metabolome, microbiome) with sociodemographic, environmental, and psychological determinants of individual pregnant people to solve the puzzle of preterm birth.
Collapse
Affiliation(s)
- David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Biomedical Innovations Building (BMI), 240 Pasteur Drive, Room 2652, Stanford, CA 94305, USA.
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Division of Reproductive, Stem Cell and Perinatal Biology, Stanford University of School of Medicine, Biomedical Innovations Building (BMI), 240 Pasteur Drive, Module 2700, Stanford, CA 94305, USA
| | - Gary M Shaw
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Biomedical Innovations Building (BMI), 240 Pasteur Drive, Room 2652, Stanford, CA 94305, USA
| | - Sarah K England
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, 425 S. Euclid Avenue, CB 8064, St. Louis, MO 63110, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Biomedical Innovations Building (BMI), 240 Pasteur Drive, Room 2652, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Akhter T, Hedeland M, Bergquist J, Ubhayasekera K, Larsson A, Byström L, Kullinger M, Skalkidou A. Elevated Plasma Levels of Arginines During Labor Among Women with Spontaneous Preterm Birth: A Prospective Cohort Study. Am J Reprod Immunol 2024; 91:e13889. [PMID: 39031744 DOI: 10.1111/aji.13889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 07/22/2024] Open
Abstract
PROBLEM Preterm birth (PTB) is a leading cause of infant mortality and morbidity. The pathogenesis of PTB is complex and involves many factors, including socioeconomy, inflammation and infection. Asymmetric dimethylarginine, ADMA and symmetric dimethylarginine, SDMA are involved in labor as inhibitors of nitric oxide, a known relaxant of the uterine smooth muscles. Arginines are scarcely studied in relation to PTB and we aimed to investigate arginines (ADMA, SDMA and L-arginine) in women with spontaneous PTB and term birth. METHODS OF THE STUDY The study was based on data from the population-based, prospective cohort BASIC study conducted in Uppsala County, Sweden, between September 2009 and November 2018. Arginines were analyzed by Ultra-High Performance Liquid Chromatography using plasma samples taken at the onset of labor from women with spontaneous PTB (n = 34) and term birth (n = 45). We also analyzed the inflammation markers CRP, TNF-R1 and TNF-R2 and GDF-15. RESULTS Women with spontaneous PTB had higher plasma levels of ADMA (p < 0.001), and L-Arginine (p = 0.03). In addition, inflammation marker, TNF-R1 (p = 0.01) was higher in spontaneous PTB compared to term birth. Further, in spontaneous PTB, no significant correlations could be observed when comparing levels of arginines with inflammation markers, except ADMA versus CRP. CONCLUSIONS These findings provide novel evidence for the potential involvement of arginines in the pathogenesis of spontaneous PTB and it seems that arginine levels at labor vary independently of several inflammatory markers. Further research is warranted to investigate the potential of arginines as therapeutic targets in the prevention and management of spontaneous PTB.
Collapse
Affiliation(s)
- Tansim Akhter
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Kumari Ubhayasekera
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ludvig Byström
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
| | - Merit Kullinger
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
- Center for Clinical Research, Västmanland Hospital, Västerås, Sweden
| | - Alkistis Skalkidou
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Feyaerts D, Marić I, Arck PC, Prins JR, Gomez-Lopez N, Gaudillière B, Stelzer IA. Predicting Spontaneous Preterm Birth Using the Immunome. Clin Perinatol 2024; 51:441-459. [PMID: 38705651 DOI: 10.1016/j.clp.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Throughout pregnancy, the maternal peripheral circulation contains valuable information reflecting pregnancy progression, detectable as tightly regulated immune dynamics. Local immune processes at the maternal-fetal interface and other reproductive and non-reproductive tissues are likely to be the pacemakers for this peripheral immune "clock." This cellular immune status of pregnancy can be leveraged for the early risk assessment and prediction of spontaneous preterm birth (sPTB). Systems immunology approaches to sPTB subtypes and cross-tissue (local and peripheral) interactions, as well as integration of multiple biological data modalities promise to improve our understanding of preterm birth pathobiology and identify potential clinically actionable biomarkers.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Petra C Arck
- Department of Obstetrics and Fetal Medicine and Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Postbus 30.001, 9700RB, Groningen, The Netherlands
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, 425 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Palo Alto, CA 94304, USA
| | - Ina A Stelzer
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Mirzaei A, Hiller BC, Stelzer IA, Thiele K, Tan Y, Becker M. Computational Approaches for Connecting Maternal Stress to Preterm Birth. Clin Perinatol 2024; 51:345-360. [PMID: 38705645 DOI: 10.1016/j.clp.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Multiple studies have hinted at a complex connection between maternal stress and preterm birth (PTB). This article describes the potential of computational methods to provide new insights into this relationship. For this, we outline existing approaches for stress assessments and various data modalities available for profiling stress responses, and review studies that sought either to establish a connection between stress and PTB or to predict PTB based on stress-related factors. Finally, we summarize the challenges of computational methods, highlighting potential future research directions within this field.
Collapse
Affiliation(s)
- Amin Mirzaei
- Department of Computer Science and Electrical Engineering, Institute for Visual and Analytic Computing, Universität Rostock, Albert-Einstein-Straße 22, 18059 Rostock, Germany
| | - Bjarne C Hiller
- Department of Computer Science and Electrical Engineering, Institute for Visual and Analytic Computing, Universität Rostock, Albert-Einstein-Straße 22, 18059 Rostock, Germany
| | - Ina A Stelzer
- Department of Pathology, University of California San Diego, GPL/CMM-West, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kristin Thiele
- Division for Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Center for Obstetrics and Pediatrics, Martinistrasse 52, 20246 Hamburg, Germany
| | - Yuqi Tan
- Department of Microbiology and Immunology, Stanford University School of Medicine, CSSR3220, 269 Campus Drive, Stanford, CA 94305, USA
| | - Martin Becker
- Department of Computer Science and Electrical Engineering, Institute for Visual and Analytic Computing, Universität Rostock, Albert-Einstein-Straße 22, 18059 Rostock, Germany.
| |
Collapse
|
24
|
Seong D, Espinosa C, Aghaeepour N. Computational Approaches for Predicting Preterm Birth and Newborn Outcomes. Clin Perinatol 2024; 51:461-473. [PMID: 38705652 PMCID: PMC11070639 DOI: 10.1016/j.clp.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Preterm birth (PTB) and its associated morbidities are a leading cause of infant mortality and morbidity. Accurate predictive models and a better biological understanding of PTB-associated morbidities are critical in reducing their adverse effects. Increasing availability of multimodal high-dimensional data sets with concurrent advances in artificial intelligence (AI) have created a rich opportunity to gain novel insights into PTB, a clinically complex and multifactorial disease. Here, the authors review the use of AI to analyze 3 modes of data: electronic health records, biological omics, and social determinants of health metrics.
Collapse
Affiliation(s)
- David Seong
- Immunology Program, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Medical Scientist Training Program, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA
| | - Camilo Espinosa
- Immunology Program, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Biomedical Data Science, Stanford University, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA; Department of Biomedical Data Science, Stanford University, 300 Pasteur Drive, Grant S280, Stanford, CA 94305-5117, USA.
| |
Collapse
|
25
|
Marić I, Stevenson DK, Aghaeepour N, Gaudillière B, Wong RJ, Angst MS. Predicting Preterm Birth Using Proteomics. Clin Perinatol 2024; 51:391-409. [PMID: 38705648 PMCID: PMC11186213 DOI: 10.1016/j.clp.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
The complexity of preterm birth (PTB), both spontaneous and medically indicated, and its various etiologies and associated risk factors pose a significant challenge for developing tools to accurately predict risk. This review focuses on the discovery of proteomics signatures that might be useful for predicting spontaneous PTB or preeclampsia, which often results in PTB. We describe methods for proteomics analyses, proteomics biomarker candidates that have so far been identified, obstacles for discovering biomarkers that are sufficiently accurate for clinical use, and the derivation of composite signatures including clinical parameters to increase predictive power.
Collapse
Affiliation(s)
- Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA.
| | - David K Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Grant S280, Stanford, CA 94305, USA
| | - Ronald J Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Grant Building, Office 276A, 300 Pasteur Drive, Stanford, CA 94305-5117, USA
| |
Collapse
|
26
|
Zhang G, Lin W, Gao N, Lan C, Ren M, Yan L, Pan B, Xu J, Han B, Hu L, Chen Y, Wu T, Zhuang L, Lu Q, Wang B, Fang M. Using Machine Learning to Construct the Blood-Follicle Distribution Models of Various Trace Elements and Explore the Transport-Related Pathways with Multiomics Data. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:7743-7757. [PMID: 38652822 DOI: 10.1021/acs.est.3c10904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Permeabilities of various trace elements (TEs) through the blood-follicle barrier (BFB) play an important role in oocyte development. However, it has not been comprehensively described as well as its involved biological pathways. Our study aimed to construct a blood-follicle distribution model of the concerned TEs and explore their related biological pathways. We finally included a total of 168 women from a cohort of in vitro fertilization-embryo transfer conducted in two reproductive centers in Beijing City and Shandong Province, China. The concentrations of 35 TEs in both serum and follicular fluid (FF) samples from the 168 women were measured, as well as the multiomics features of the metabolome, lipidome, and proteome in both plasma and FF samples. Multiomics features associated with the transfer efficiencies of TEs through the BFB were selected by using an elastic net model and further utilized for pathway analysis. Various machine learning (ML) models were built to predict the concentrations of TEs in FF. Overall, there are 21 TEs that exhibited three types of consistent BFB distribution characteristics between Beijing and Shandong centers. Among them, the concentrations of arsenic, manganese, nickel, tin, and bismuth in FF were higher than those in the serum with transfer efficiencies of 1.19-4.38, while a reverse trend was observed for the 15 TEs with transfer efficiencies of 0.076-0.905, e.g., mercury, germanium, selenium, antimony, and titanium. Lastly, cadmium was evenly distributed in the two compartments with transfer efficiencies of 0.998-1.056. Multiomics analysis showed that the enrichment of TEs was associated with the synthesis and action of steroid hormones and the glucose metabolism. Random forest model can provide the most accurate predictions of the concentrations of TEs in FF among the concerned ML models. In conclusion, the selective permeability through the BFB for various TEs may be significantly regulated by the steroid hormones and the glucose metabolism. Also, the concentrations of some TEs in FF can be well predicted by their serum levels with a random forest model.
Collapse
Affiliation(s)
- Guohuan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Weinan Lin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Ning Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Changxin Lan
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Mengyuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Lailai Yan
- Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing 100191, P. R. China
| | - Bo Pan
- Yunnan Provincial Key Lab of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming 650500, P. R. China
| | - Jia Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| | - Bin Han
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, P. R. China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Science, Chinese Academy of Sciences, Beijing 100085, P. R. China
| | - Yuanchen Chen
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, P. R. China
| | - Tianxiang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
| | - Lili Zhuang
- Reproductive Medicine Centre, Yuhuangding Hospital of Yantai, Affiliated Hospital of Qingdao University, Yantai 264000, P. R. China
| | - Qun Lu
- Medical Center for Human Reproduction, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R China
- Center of Reproductive Medicine, Peking University People's Hospital, Beijing 100044, P. R. China
| | - Bin Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, P. R. China
- Institute of Reproductive and Child Health, School of Public Health, Peking University, Beijing 100191, P. R. China
- Key Laboratory of Reproductive Health, National Health and Family Planning Commission of the People's Republic of China, Beijing 100191, P. R. China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, P. R. China
- Laboratory for Earth Surface Processes, College of Urban and Environmental Science, Peking University, Beijing 100871, China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
27
|
Ho SJ, Chaput D, Sinkey RG, Garces AH, New EP, Okuka M, Sang P, Arlier S, Semerci N, Steffensen TS, Rutherford TJ, Alsina AE, Cai J, Anderson ML, Magness RR, Uversky VN, Cummings DAT, Tsibris JCM. Proteomic studies of VEGFR2 in human placentas reveal protein associations with preeclampsia, diabetes, gravidity, and labor. Cell Commun Signal 2024; 22:221. [PMID: 38594674 PMCID: PMC11003095 DOI: 10.1186/s12964-024-01567-0] [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: 10/21/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
VEGFR2 (Vascular endothelial growth factor receptor 2) is a central regulator of placental angiogenesis. The study of the VEGFR2 proteome of chorionic villi at term revealed its partners MDMX (Double minute 4 protein) and PICALM (Phosphatidylinositol-binding clathrin assembly protein). Subsequently, the oxytocin receptor (OT-R) and vasopressin V1aR receptor were detected in MDMX and PICALM immunoprecipitations. Immunogold electron microscopy showed VEGFR2 on endothelial cell (EC) nuclei, mitochondria, and Hofbauer cells (HC), tissue-resident macrophages of the placenta. MDMX, PICALM, and V1aR were located on EC plasma membranes, nuclei, and HC nuclei. Unexpectedly, PICALM and OT-R were detected on EC projections into the fetal lumen and OT-R on 20-150 nm clusters therein, prompting the hypothesis that placental exosomes transport OT-R to the fetus and across the blood-brain barrier. Insights on gestational complications were gained by univariable and multivariable regression analyses associating preeclampsia with lower MDMX protein levels in membrane extracts of chorionic villi, and lower MDMX, PICALM, OT-R, and V1aR with spontaneous vaginal deliveries compared to cesarean deliveries before the onset of labor. We found select associations between higher MDMX, PICALM, OT-R protein levels and either gravidity, diabetes, BMI, maternal age, or neonatal weight, and correlations only between PICALM-OT-R (p < 2.7 × 10-8), PICALM-V1aR (p < 0.006), and OT-R-V1aR (p < 0.001). These results offer for exploration new partnerships in metabolic networks, tissue-resident immunity, and labor, notably for HC that predominantly express MDMX.
Collapse
Grants
- Department of Obstetrics and Gynecology, University of South Florida
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida
- Department of Chemistry, University of South Florida
- Tampa General Hospital, Tampa, Florida
- Teasley Foundation
- Department of Molecular Medicine, University of South Florida
- Department of Biology, University of Florida
- Emerging Pathogens Institute, University of Florida
Collapse
Affiliation(s)
- Shannon J Ho
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Amanda H Garces
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida, Tampa, FL, USA
| | - Erika P New
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Sefa Arlier
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | | | - Thomas J Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Angel E Alsina
- Transplant Surgery Center, Tampa General Hospital, Tampa, FL, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Matthew L Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Derek A T Cummings
- Department of Biology and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - John C M Tsibris
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA.
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
28
|
Zhu B, Bai Y, Yeo YY, Lu X, Rovira-Clavé X, Chen H, Yeung J, Gerber GK, Angelo M, Shalek AK, Nolan GP, Jiang S. A Spatial Multi-Modal Dissection of Host-Microbiome Interactions within the Colitis Tissue Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583400. [PMID: 38496402 PMCID: PMC10942342 DOI: 10.1101/2024.03.04.583400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The intricate and dynamic interactions between the host immune system and its microbiome constituents undergo dynamic shifts in response to perturbations to the intestinal tissue environment. Our ability to study these events on the systems level is significantly limited by in situ approaches capable of generating simultaneous insights from both host and microbial communities. Here, we introduce Microbiome Cartography (MicroCart), a framework for simultaneous in situ probing of host features and its microbiome across multiple spatial modalities. We demonstrate MicroCart by comprehensively investigating the alterations in both gut host and microbiome components in a murine model of colitis by coupling MicroCart with spatial proteomics, transcriptomics, and glycomics platforms. Our findings reveal a global but systematic transformation in tissue immune responses, encompassing tissue-level remodeling in response to host immune and epithelial cell state perturbations, and bacterial population shifts, localized inflammatory responses, and metabolic process alterations during colitis. MicroCart enables a deep investigation of the intricate interplay between the host tissue and its microbiome with spatial multiomics.
Collapse
Affiliation(s)
- Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Yunhao Bai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xiaowei Lu
- Mass Spectrometry Core Facility, Stanford University, Stanford, CA, United States
| | - Xavier Rovira-Clavé
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Biological and Medical Informatics program, UCSF, San Francisco, CA, United States
| | - Jason Yeung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Georg K Gerber
- Division of Computational Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Health Sciences and Technology, Harvard University and MIT, Cambridge, MA, USA
| | - Mike Angelo
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Sizun Jiang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Mallick H, Porwal A, Saha S, Basak P, Svetnik V, Paul E. An integrated Bayesian framework for multi-omics prediction and classification. Stat Med 2024; 43:983-1002. [PMID: 38146838 DOI: 10.1002/sim.9953] [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: 04/21/2022] [Revised: 10/06/2023] [Accepted: 10/24/2023] [Indexed: 12/27/2023]
Abstract
With the growing commonality of multi-omics datasets, there is now increasing evidence that integrated omics profiles lead to more efficient discovery of clinically actionable biomarkers that enable better disease outcome prediction and patient stratification. Several methods exist to perform host phenotype prediction from cross-sectional, single-omics data modalities but decentralized frameworks that jointly analyze multiple time-dependent omics data to highlight the integrative and dynamic impact of repeatedly measured biomarkers are currently limited. In this article, we propose a novel Bayesian ensemble method to consolidate prediction by combining information across several longitudinal and cross-sectional omics data layers. Unlike existing frequentist paradigms, our approach enables uncertainty quantification in prediction as well as interval estimation for a variety of quantities of interest based on posterior summaries. We apply our method to four published multi-omics datasets and demonstrate that it recapitulates known biology in addition to providing novel insights while also outperforming existing methods in estimation, prediction, and uncertainty quantification. Our open-source software is publicly available at https://github.com/himelmallick/IntegratedLearner.
Collapse
Affiliation(s)
- Himel Mallick
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, 10065, New York, USA
- Department of Statistics and Data Science, Cornell University, Ithaca, New York, USA
| | - Anupreet Porwal
- Department of Statistics, University of Washington, Seattle, Washington, USA
| | - Satabdi Saha
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Piyali Basak
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Vladimir Svetnik
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Erina Paul
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, New Jersey, USA
| |
Collapse
|
30
|
Basavaraj C, Grant AD, Aras SG, Erickson EN. Deep Learning Model Using Continuous Skin Temperature Data Predicts Labor Onset. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.25.24303344. [PMID: 38464102 PMCID: PMC10925356 DOI: 10.1101/2024.02.25.24303344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Changes in body temperature anticipate labor onset in numerous mammals, yet this concept has not been explored in humans. Methods We evaluated patterns in continuous skin temperature data in 91 pregnant women using a wearable smart ring. Additionally, we collected daily steroid hormone samples leading up to labor in a subset of 28 pregnancies and analyzed relationships among hormones and body temperature trajectory. Finally, we developed a novel autoencoder long-short-term-memory (AE-LSTM) deep learning model to provide a daily estimation of days until labor onset. Results Features of temperature change leading up to labor were associated with urinary hormones and labor type. Spontaneous labors exhibited greater estriol to α-pregnanediol ratio, as well as lower body temperature and more stable circadian rhythms compared to pregnancies that did not undergo spontaneous labor. Skin temperature data from 54 pregnancies that underwent spontaneous labor between 34 and 42 weeks of gestation were included in training the AE-LSTM model, and an additional 40 pregnancies that underwent artificial induction of labor or Cesarean without labor were used for further testing. The model was trained only on aggregate 5-minute skin temperature data starting at a gestational age of 240 until labor onset. During cross-validation AE-LSTM average error (true - predicted) dropped below 2 days at 8 days before labor, independent of gestational age. Labor onset windows were calculated from the AE-LSTM output using a probabilistic distribution of model error. For these windows AE-LSTM correctly predicted labor start for 79% of the spontaneous labors within a 4.6-day window at 7 days before true labor, and 7.4-day window at 10 days before true labor. Conclusion Continuous skin temperature reflects progression toward labor and hormonal status during pregnancy. Deep learning using continuous temperature may provide clinically valuable tools for pregnancy care.
Collapse
Affiliation(s)
- Chinmai Basavaraj
- Department of Computer Science, The University of Arizona, Tucson, AZ, USA
| | | | - Shravan G Aras
- Center for Biomedical Informatics and Biostatistics, The University of Arizona Health Sciences, Tucson, AZ, USA
| | | |
Collapse
|
31
|
Gondane P, Kumbhakarn S, Maity P, Kapat K. Recent Advances and Challenges in the Early Diagnosis and Treatment of Preterm Labor. Bioengineering (Basel) 2024; 11:161. [PMID: 38391647 PMCID: PMC10886370 DOI: 10.3390/bioengineering11020161] [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: 12/30/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Preterm birth (PTB) is the primary cause of neonatal mortality and long-term disabilities. The unknown mechanism behind PTB makes diagnosis difficult, yet early detection is necessary for controlling and averting related consequences. The primary focus of this work is to provide an overview of the known risk factors associated with preterm labor and the conventional and advanced procedures for early detection of PTB, including multi-omics and artificial intelligence/machine learning (AI/ML)- based approaches. It also discusses the principles of detecting various proteomic biomarkers based on lateral flow immunoassay and microfluidic chips, along with the commercially available point-of-care testing (POCT) devices and associated challenges. After briefing the therapeutic and preventive measures of PTB, this review summarizes with an outlook.
Collapse
Affiliation(s)
- Prashil Gondane
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, India
| | - Sakshi Kumbhakarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, India
| | - Pritiprasanna Maity
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, India
| |
Collapse
|
32
|
Becker M, Fehr K, Goguen S, Miliku K, Field C, Robertson B, Yonemitsu C, Bode L, Simons E, Marshall J, Dawod B, Mandhane P, Turvey SE, Moraes TJ, Subbarao P, Rodriguez N, Aghaeepour N, Azad MB. Multimodal machine learning for modeling infant head circumference, mothers' milk composition, and their shared environment. Sci Rep 2024; 14:2977. [PMID: 38316895 PMCID: PMC10844250 DOI: 10.1038/s41598-024-52323-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: 09/28/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024] Open
Abstract
Links between human milk (HM) and infant development are poorly understood and often focus on individual HM components. Here we apply multi-modal predictive machine learning to study HM and head circumference (a proxy for brain development) among 1022 mother-infant dyads of the CHILD Cohort. We integrated HM data (19 oligosaccharides, 28 fatty acids, 3 hormones, 28 chemokines) with maternal and infant demographic, health, dietary and home environment data. Head circumference was significantly predictable at 3 and 12 months. Two of the most associated features were HM n3-polyunsaturated fatty acid C22:6n3 (docosahexaenoic acid, DHA; p = 9.6e-05) and maternal intake of fish (p = 4.1e-03), a key dietary source of DHA with established relationships to brain function. Thus, using a systems biology approach, we identified meaningful relationships between HM and brain development, which validates our statistical approach, gives credence to the novel associations we observed, and sets the foundation for further research with additional cohorts and HM analytes.
Collapse
Affiliation(s)
- Martin Becker
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada
- Stanford University, Stanford, 94305, USA
| | - Kelsey Fehr
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Stephanie Goguen
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Kozeta Miliku
- University of Toronto, Toronto, M5S 1A8, Canada
- McMaster University, Hamilton, M5S 1A8, Canada
| | | | | | - Chloe Yonemitsu
- University of California, San Diego, La Jolla, CA, 92093, USA
| | - Lars Bode
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada
- University of California, San Diego, La Jolla, CA, 92093, USA
| | | | | | | | | | - Stuart E Turvey
- University of British Columbia and British Columbia Children's Hospital, Vancouver, V5Z4H4, Canada
| | | | - Padmaja Subbarao
- University of Toronto, Toronto, M5S 1A8, Canada
- McMaster University, Hamilton, M5S 1A8, Canada
- SickKids, Toronto, M5G 0A4, Canada
| | - Natalie Rodriguez
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
- University of Manitoba, Winnipeg, R3E3P4, Canada
| | - Nima Aghaeepour
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada.
- Stanford University, Stanford, 94305, USA.
| | - Meghan B Azad
- International Milk Composition (IMiC) Consortium, Winnipeg, Canada.
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, Canada.
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
- University of Manitoba, Winnipeg, R3E3P4, Canada.
| |
Collapse
|
33
|
Golob JL, Oskotsky TT, Tang AS, Roldan A, Chung V, Ha CWY, Wong RJ, Flynn KJ, Parraga-Leo A, Wibrand C, Minot SS, Oskotsky B, Andreoletti G, Kosti I, Bletz J, Nelson A, Gao J, Wei Z, Chen G, Tang ZZ, Novielli P, Romano D, Pantaleo E, Amoroso N, Monaco A, Vacca M, De Angelis M, Bellotti R, Tangaro S, Kuntzleman A, Bigcraft I, Techtmann S, Bae D, Kim E, Jeon J, Joe S, Theis KR, Ng S, Lee YS, Diaz-Gimeno P, Bennett PR, MacIntyre DA, Stolovitzky G, Lynch SV, Albrecht J, Gomez-Lopez N, Romero R, Stevenson DK, Aghaeepour N, Tarca AL, Costello JC, Sirota M. Microbiome preterm birth DREAM challenge: Crowdsourcing machine learning approaches to advance preterm birth research. Cell Rep Med 2024; 5:101350. [PMID: 38134931 PMCID: PMC10829755 DOI: 10.1016/j.xcrm.2023.101350] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/15/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
Every year, 11% of infants are born preterm with significant health consequences, with the vaginal microbiome a risk factor for preterm birth. We crowdsource models to predict (1) preterm birth (PTB; <37 weeks) or (2) early preterm birth (ePTB; <32 weeks) from 9 vaginal microbiome studies representing 3,578 samples from 1,268 pregnant individuals, aggregated from public raw data via phylogenetic harmonization. The predictive models are validated on two independent unpublished datasets representing 331 samples from 148 pregnant individuals. The top-performing models (among 148 and 121 submissions from 318 teams) achieve area under the receiver operator characteristic (AUROC) curve scores of 0.69 and 0.87 predicting PTB and ePTB, respectively. Alpha diversity, VALENCIA community state types, and composition are important features in the top-performing models, most of which are tree-based methods. This work is a model for translation of microbiome data into clinically relevant predictive models and to better understand preterm birth.
Collapse
Affiliation(s)
- Jonathan L Golob
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA.
| | - Tomiko T Oskotsky
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| | - Alice S Tang
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Alennie Roldan
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | | | - Connie W Y Ha
- Benioff Center for Microbiome Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; March of Dimes Prematurity Research Center at Stanford University, Stanford, CA, USA
| | | | - Antonio Parraga-Leo
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, Obstetrics and Gynaecology, Universidad de Valencia, Valencia, Spain; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Camilla Wibrand
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Samuel S Minot
- Data Core, Shared Resources, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Boris Oskotsky
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
| | - Gaia Andreoletti
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Idit Kosti
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Jifan Gao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhoujingpeng Wei
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Guanhua Chen
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zheng-Zheng Tang
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Pierfrancesco Novielli
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Bari, Italy; Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy
| | - Donato Romano
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Bari, Italy; Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy
| | - Ester Pantaleo
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy; Dipartimento Interateneo di Fisica "M, Merlin", Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Nicola Amoroso
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy; Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy; Dipartimento Interateneo di Fisica "M, Merlin", Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Mirco Vacca
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Maria De Angelis
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Roberto Bellotti
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy; Dipartimento Interateneo di Fisica "M, Merlin", Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Sabina Tangaro
- Dipartimento di Scienze del Suolo, della Pianta e degli Alimenti, Università degli Studi di Bari Aldo Moro, Bari, Italy; Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Bari, Italy
| | - Abigail Kuntzleman
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Isaac Bigcraft
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Stephen Techtmann
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, USA
| | - Daehun Bae
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Eunyoung Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jongbum Jeon
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Soobok Joe
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kevin R Theis
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, USA
| | - Sherrianne Ng
- Imperial College Parturition Research Group, Division of the Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK
| | - Yun S Lee
- Imperial College Parturition Research Group, Division of the Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK
| | - Patricia Diaz-Gimeno
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Phillip R Bennett
- Imperial College Parturition Research Group, Division of the Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK
| | - David A MacIntyre
- Imperial College Parturition Research Group, Division of the Institute of Reproductive and Developmental Biology, Imperial College London, London, UK; March of Dimes Prematurity Research Centre at Imperial College London, London, UK
| | - Gustavo Stolovitzky
- Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA; Thomas J. Watson Research Center, IBM, Yorktown Heights, NY, USA; Sema4, Stamford, CT, USA
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Nardhy Gomez-Lopez
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, USA; Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA; Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Center for Academic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - James C Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marina Sirota
- March of Dimes Prematurity Research Center at the University of California San Francisco, San Francisco, CA, USA; Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Garcia-Flores V, Romero R, Tarca AL, Peyvandipour A, Xu Y, Galaz J, Miller D, Chaiworapongsa T, Chaemsaithong P, Berry SM, Awonuga AO, Bryant DR, Pique-Regi R, Gomez-Lopez N. Deciphering maternal-fetal cross-talk in the human placenta during parturition using single-cell RNA sequencing. Sci Transl Med 2024; 16:eadh8335. [PMID: 38198568 PMCID: PMC11238316 DOI: 10.1126/scitranslmed.adh8335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Labor is a complex physiological process requiring a well-orchestrated dialogue between the mother and fetus. However, the cellular contributions and communications that facilitate maternal-fetal cross-talk in labor have not been fully elucidated. Here, single-cell RNA sequencing (scRNA-seq) was applied to decipher maternal-fetal signaling in the human placenta during term labor. First, a single-cell atlas of the human placenta was established, demonstrating that maternal and fetal cell types underwent changes in transcriptomic activity during labor. Cell types most affected by labor were fetal stromal and maternal decidual cells in the chorioamniotic membranes (CAMs) and maternal and fetal myeloid cells in the placenta. Cell-cell interaction analyses showed that CAM and placental cell types participated in labor-driven maternal and fetal signaling, including the collagen, C-X-C motif ligand (CXCL), tumor necrosis factor (TNF), galectin, and interleukin-6 (IL-6) pathways. Integration of scRNA-seq data with publicly available bulk transcriptomic data showed that placenta-derived scRNA-seq signatures could be monitored in the maternal circulation throughout gestation and in labor. Moreover, comparative analysis revealed that placenta-derived signatures in term labor were mirrored by those in spontaneous preterm labor and birth. Furthermore, we demonstrated that early in gestation, labor-specific, placenta-derived signatures could be detected in the circulation of women destined to undergo spontaneous preterm birth, with either intact or prelabor ruptured membranes. Collectively, our findings provide insight into the maternal-fetal cross-talk of human parturition and suggest that placenta-derived single-cell signatures can aid in the development of noninvasive biomarkers for the prediction of preterm birth.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48201, USA
| | - Azam Peyvandipour
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Piya Chaemsaithong
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Stanley M Berry
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Awoniyi O Awonuga
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - David R Bryant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
35
|
Lian X, Zhang Y, Zhou Y, Sun X, Huang S, Dai H, Han L, Zhu F. SingPro: a knowledge base providing single-cell proteomic data. Nucleic Acids Res 2024; 52:D552-D561. [PMID: 37819028 PMCID: PMC10767818 DOI: 10.1093/nar/gkad830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/03/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023] Open
Abstract
Single-cell proteomics (SCP) has emerged as a powerful tool for detecting cellular heterogeneity, offering unprecedented insights into biological mechanisms that are masked in bulk cell populations. With the rapid advancements in AI-based time trajectory analysis and cell subpopulation identification, there exists a pressing need for a database that not only provides SCP raw data but also explicitly describes experimental details and protein expression profiles. However, no such database has been available yet. In this study, a database, entitled 'SingPro', specializing in single-cell proteomics was thus developed. It was unique in (a) systematically providing the SCP raw data for both mass spectrometry-based and flow cytometry-based studies and (b) explicitly describing experimental detail for SCP study and expression profile of any studied protein. Anticipating a robust interest from the research community, this database is poised to become an invaluable repository for OMICs-based biomedical studies. Access to SingPro is unrestricted and does not mandate a login at: http://idrblab.org/singpro/.
Collapse
Affiliation(s)
- Xichen Lian
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai 315211, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shijie Huang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Haibin Dai
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lianyi Han
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai 315211, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
36
|
Shcherbina M, Potapova L, Lipko O, Shcherbina I, Mertsalova O. Association of the key immunological and hemodynamic determinants with cervix ripening in pregnant women. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:201-207. [PMID: 38592979 DOI: 10.36740/wlek202402103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
OBJECTIVE Aim: To investigate a correlation between cervical ripening, the immunological features and the hemodynamic characteristics of the cervix during the preparation for vaginal labor. PATIENTS AND METHODS Materials and Methods: We examined 75 pregnant women at different gestational age. General clinical and immunological studies were conducted in order to check serum concentration of cytokines IL-6, IL-1β, and TNF-α. Ultrasound and Doppler study were used to determine resistance index and systolic-diastolic ratio of blood flow in the common uterine artery as well as the descending and ascending parts and cervical stromal arteries. RESULTS Results: Pregnant women with high cervical ripening score had high concentrations of the major proinflammatory cytokines (IL-1β, IL-6, and TNF-α). Analysis of the of the cervical blood flow indicators of the studied groups showed significant differences in the indices of vascular resistance in the vessels that feed the cervix. Our data showed a significant correlation between the cervix ripening and both the serum levels of the studied cytokines and the level of peripheral vascular resistance indices in the common uterine arteries of the cervix, and the blood flow indices in the cervical stromal vessels. CONCLUSION Conclusions: Our study shows that the process of preparing the woman's body for labor is associated with immunological adjustment and increased hemodynamics of the cervix. We report that cervical ripening is associated with the immunological components and hemodynamic parameters of the cervix at late-stage pregnancy. Measuring cervix ripening and the accompanied changes in cytokine levels and hemodynamic parameters will form a more accurate assessment of birth preparedness and labor complications.
Collapse
Affiliation(s)
| | | | - Oksana Lipko
- KHARKIV NATIONAL MEDICAL UNIVERSITY, KHARKIV, UKRAINE
| | | | | |
Collapse
|
37
|
Ghazvini S, Uthaman S, Synan L, Lin EC, Sarkar S, Santillan MK, Santillan DA, Bardhan R. Predicting the onset of preeclampsia by longitudinal monitoring of metabolic changes throughout pregnancy with Raman spectroscopy. Bioeng Transl Med 2024; 9:e10595. [PMID: 38193120 PMCID: PMC10771567 DOI: 10.1002/btm2.10595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2023] [Accepted: 08/15/2023] [Indexed: 01/10/2024] Open
Abstract
Preeclampsia is a life-threatening pregnancy disorder. Current clinical assays cannot predict the onset of preeclampsia until the late 2nd trimester, which often leads to poor maternal and neonatal outcomes. Here we show that Raman spectroscopy combined with machine learning in pregnant patient plasma enables rapid, highly sensitive maternal metabolome screening that predicts preeclampsia as early as the 1st trimester with >82% accuracy. We identified 12, 15 and 17 statistically significant metabolites in the 1st, 2nd and 3rd trimesters, respectively. Metabolic pathway analysis shows multiple pathways corresponding to amino acids, fatty acids, retinol, and sugars are enriched in the preeclamptic cohort relative to a healthy pregnancy. Leveraging Pearson's correlation analysis, we show for the first time with Raman Spectroscopy that metabolites are associated with several clinical factors, including patients' body mass index, gestational age at delivery, history of preeclampsia, and severity of preeclampsia. We also show that protein quantification alone of proinflammatory cytokines and clinically relevant angiogenic markers are inadequate in identifying at-risk patients. Our findings demonstrate that Raman spectroscopy is a powerful tool that may complement current clinical assays in early diagnosis and in the prognosis of the severity of preeclampsia to ultimately enable comprehensive prenatal care for all patients.
Collapse
Affiliation(s)
- Saman Ghazvini
- Department of Chemical and Biological EngineeringIowa State UniversityAmesIowaUSA
- Nanovaccine InstituteIowa State UniversityAmesIowaUSA
| | - Saji Uthaman
- Department of Chemical and Biological EngineeringIowa State UniversityAmesIowaUSA
- Nanovaccine InstituteIowa State UniversityAmesIowaUSA
| | - Lilly Synan
- Department of Chemical and Biological EngineeringIowa State UniversityAmesIowaUSA
- Nanovaccine InstituteIowa State UniversityAmesIowaUSA
| | - Eugene C. Lin
- Department of Chemistry and BiochemistryNational Chung Cheng UniversityChiayiTaiwan
| | - Soumik Sarkar
- Department of Mechanical EngineeringIowa state UniversityAmesIowaUSA
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, Carver College of MedicineUniversity of Iowa, Hospitals & ClinicsIowa CityIowaUSA
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, Carver College of MedicineUniversity of Iowa, Hospitals & ClinicsIowa CityIowaUSA
| | - Rizia Bardhan
- Department of Chemical and Biological EngineeringIowa State UniversityAmesIowaUSA
- Nanovaccine InstituteIowa State UniversityAmesIowaUSA
| |
Collapse
|
38
|
Mengelkoch S, Gassen J, Lev-Ari S, Alley JC, Schüssler-Fiorenza Rose SM, Snyder MP, Slavich GM. Multi-omics in stress and health research: study designs that will drive the field forward. Stress 2024; 27:2321610. [PMID: 38425100 PMCID: PMC11216062 DOI: 10.1080/10253890.2024.2321610] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Despite decades of stress research, there still exist substantial gaps in our understanding of how social, environmental, and biological factors interact and combine with developmental stressor exposures, cognitive appraisals of stressors, and psychosocial coping processes to shape individuals' stress reactivity, health, and disease risk. Relatively new biological profiling approaches, called multi-omics, are helping address these issues by enabling researchers to quantify thousands of molecules from a single blood or tissue sample, thus providing a panoramic snapshot of the molecular processes occurring in an organism from a systems perspective. In this review, we summarize two types of research designs for which multi-omics approaches are best suited, and describe how these approaches can help advance our understanding of stress processes and the development, prevention, and treatment of stress-related pathologies. We first discuss incorporating multi-omics approaches into theory-rich, intensive longitudinal study designs to characterize, in high-resolution, the transition to stress-related multisystem dysfunction and disease throughout development. Next, we discuss how multi-omics approaches should be incorporated into intervention research to better understand the transition from stress-related dysfunction back to health, which can help inform novel precision medicine approaches to managing stress and fostering biopsychosocial resilience. Throughout, we provide concrete recommendations for types of studies that will help advance stress research, and translate multi-omics data into better health and health care.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Jeffrey Gassen
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Shahar Lev-Ari
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Health Promotion, Tel Aviv University, Tel Aviv, Israel
| | - Jenna C. Alley
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | | | | | - George M. Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Galaz J, Romero R, Greenberg JM, Theis KR, Arenas-Hernandez M, Xu Y, Farias-Jofre M, Miller D, Kanninen T, Garcia-Flores V, Gomez-Lopez N. Host-microbiome interactions in distinct subsets of preterm labor and birth. iScience 2023; 26:108341. [PMID: 38047079 PMCID: PMC10692673 DOI: 10.1016/j.isci.2023.108341] [Citation(s) in RCA: 4] [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: 05/30/2023] [Revised: 09/06/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Preterm birth, the leading cause of perinatal morbidity, often follows premature labor, a syndrome whose prevention remains a challenge. To better understand the relationship between premature labor and host-microbiome interactions, we conducted a mechanistic investigation using three preterm birth models. We report that intra-amniotic delivery of LPS triggers inflammatory responses in the amniotic cavity and cervico-vaginal microenvironment, causing vaginal microbiome changes and signs of active labor. Intra-amniotic IL-1α delivery causes a moderate inflammatory response in the amniotic cavity but increasing inflammation in the cervico-vaginal space, leading to vaginal microbiome disruption and signs of active labor. Conversely, progesterone action blockade by RU-486 triggers local immune responses accompanying signs of active labor without altering the vaginal microbiome. Preterm labor facilitates ascension of cervico-vaginal bacteria into the amniotic cavity, regardless of stimulus. This study provides compelling mechanistic insights into the dynamic host-microbiome interactions within the cervico-vaginal microenvironment that accompany premature labor and birth.
Collapse
Affiliation(s)
- Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Jonathan M. Greenberg
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kevin R. Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
40
|
Tu WB, Christofk HR, Plath K. Nutrient regulation of development and cell fate decisions. Development 2023; 150:dev199961. [PMID: 37260407 PMCID: PMC10281554 DOI: 10.1242/dev.199961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Diet contributes to health at all stages of life, from embryonic development to old age. Nutrients, including vitamins, amino acids, lipids and sugars, have instructive roles in directing cell fate and function, maintaining stem cell populations, tissue homeostasis and alleviating the consequences of aging. This Review highlights recent findings that illuminate how common diets and specific nutrients impact cell fate decisions in healthy and disease contexts. We also draw attention to new models, technologies and resources that help to address outstanding questions in this emerging field and may lead to dietary approaches that promote healthy development and improve disease treatments.
Collapse
Affiliation(s)
- William B. Tu
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Heather R. Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Akhter T, Hedeland M, Bergquist J, Ubhayasekera K, Larsson A, Kullinger M, Skalkidou A. Plasma levels of arginines at term pregnancy in relation to mode of onset of labor and mode of childbirth. Am J Reprod Immunol 2023; 90:e13767. [PMID: 37641379 DOI: 10.1111/aji.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/16/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
PROBLEM The exact biochemical mechanisms that initiate labor are not yet fully understood. Nitric oxide is a potent relaxant of uterine smooth muscles until labor starts, and its precursor is L-arginine. Asymmetric (ADMA) and symmetric (SDMA) dimethylarginines, are potent NO-inhibitors. However, arginines (dimethylarginines and L-arginine) are scarcely studied in relation to labor and childbirth. We aimed to investigate arginines in women with spontaneous (SLVB) and induced (ILVB) term labor with vaginal birth and in women undergoing elective caesarean section (ECS). METHOD OF STUDY Women at gestational week 16-18 were recruited to the population-based prospective cohort study BASIC at the Uppsala University Hospital, Sweden. Plasma samples taken at start of labor were analyzed for arginines, from SLVB (n = 45), ILVB (n = 45), and ECS (n = 45), using Ultra-High Performance Liquid Chromatography. Between-group differences were assessed using Kruskal-Wallis and Mann-Whitney U-test. RESULTS Women with SLVB and ILVB had higher levels of ADMA (p < .0001), SDMA (p < .05) and lower L-arginines (p < .01), L-arginine/ADMA (p < .0001), and L-arginine/SDMA (p < .01, respectively <.001) compared to ECS. However, ILVB had higher ADMA (p < .0001) and lower L-arginine (p < .01), L-arginine/ADMA (p < .0001), and L-arginine/SDMA (p < .01) compared to SLVB. Results are adjusted for gestational length at birth and cervical dilatation at sampling. CONCLUSION Our novel findings of higher levels of dimethylarginines in term vaginal births compared to ECS give insights into the biochemical mechanisms of labor. These findings might also serve as a basis for further studies of arginines in complicated pregnancies and labor.
Collapse
Affiliation(s)
- Tansim Akhter
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
| | - Mikael Hedeland
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Medical Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, Uppsala, Sweden
| | - Kumari Ubhayasekera
- Department of Medical Chemistry, Analytical Pharmaceutical Chemistry, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Merit Kullinger
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
- Center for Clinical Research, Västerås Västmanland Hospital, Västerås, Sweden
| | - Alkistis Skalkidou
- Department of Women's and Children's Health, Section of Obstetrics and Gynecology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
42
|
Chen Y, Chen H, Zheng Q. Siglecs family used by pathogens for immune escape may engaged in immune tolerance in pregnancy. J Reprod Immunol 2023; 159:104127. [PMID: 37572430 DOI: 10.1016/j.jri.2023.104127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The Siglecs family is a group of type I sialic acid-binding immunoglobulin-like receptors that regulate cellular signaling by recognizing sialic acid epitopes. Siglecs are predominantly expressed on the surface of leukocytes, where they play a crucial role in regulating immune activity. Pathogens can exploit inhibitory Siglecs by utilizing their sialic acid components to promote invasion or suppress immune functions, facilitating immune evasion. The establishing of an immune-balanced maternal-fetal interface microenvironment is essential for a successful pregnancy. Dysfunctional immune cells may lead to adverse pregnancy outcomes. Siglecs are important for inducing a phenotypic switch in leukocytes at the maternal-fetal interface toward a less toxic and more tolerant phenotype. Recent discoveries regarding Siglecs in the reproductive system have drawn further attention to their potential roles in reproduction. In this review, we primarily discuss the latest advances in understanding the impact of Siglecs as immune regulators on infections and pregnancy.
Collapse
Affiliation(s)
- Ying Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Huan Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China
| | - Qingliang Zheng
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen 518033, PR China.
| |
Collapse
|
43
|
Erickson EN, Gotlieb N, Pereira LM, Myatt L, Mosquera-Lopez C, Jacobs PG. Predicting labor onset relative to the estimated date of delivery using smart ring physiological data. NPJ Digit Med 2023; 6:153. [PMID: 37598232 PMCID: PMC10439919 DOI: 10.1038/s41746-023-00902-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
The transition from pregnancy into parturition is physiologically directed by maternal, fetal and placental tissues. We hypothesize that these processes may be reflected in maternal physiological metrics. We enrolled pregnant participants in the third-trimester (n = 118) to study continuously worn smart ring devices monitoring heart rate, heart rate variability, skin temperature, sleep and physical activity from negative temperature coefficient, 3-D accelerometer and infrared photoplethysmography sensors. Weekly surveys assessed labor symptoms, pain, fatigue and mood. We estimated the association between each metric, gestational age, and the likelihood of a participant's labor beginning prior to (versus after) the clinical estimated delivery date (EDD) of 40.0 weeks with mixed effects regression. A boosted random forest was trained on the physiological metrics to predict pregnancies that naturally passed the EDD versus undergoing onset of labor prior to the EDD. Here we report that many raw sleep, activity, pain, fatigue and labor symptom metrics are correlated with gestational age. As gestational age advances, pregnant individuals have lower resting heart rate 0.357 beats/minute/week, 0.84 higher heart rate variability (milliseconds) and shorter durations of physical activity and sleep. Further, random forest predictions determine pregnancies that would pass the EDD with accuracy of 0.71 (area under the receiver operating curve). Self-reported symptoms of labor correlate with increased gestational age and not with the timing of labor (relative to EDD) or onset of spontaneous labor. The use of maternal smart ring-derived physiological data in the third-trimester may improve prediction of the natural duration of pregnancy relative to the EDD.
Collapse
Affiliation(s)
- Elise N Erickson
- College of Nursing / College of Pharmacy, The University of Arizona, Tucson, AZ, USA.
- Midwifery Division, School of Nursing, Oregon Health & Science University, Portland, OR, USA.
| | | | - Leonardo M Pereira
- Department of Obstetrics & Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of Obstetrics & Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Clara Mosquera-Lopez
- Artificial Intelligence for Medical Systems (AIMS) Lab, Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Peter G Jacobs
- Artificial Intelligence for Medical Systems (AIMS) Lab, Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
44
|
Moufarrej MN, Bianchi DW, Shaw GM, Stevenson DK, Quake SR. Noninvasive Prenatal Testing Using Circulating DNA and RNA: Advances, Challenges, and Possibilities. Annu Rev Biomed Data Sci 2023; 6:397-418. [PMID: 37196360 PMCID: PMC10528197 DOI: 10.1146/annurev-biodatasci-020722-094144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Prenatal screening using sequencing of circulating cell-free DNA has transformed obstetric care over the past decade and significantly reduced the number of invasive diagnostic procedures like amniocentesis for genetic disorders. Nonetheless, emergency care remains the only option for complications like preeclampsia and preterm birth, two of the most prevalent obstetrical syndromes. Advances in noninvasive prenatal testing expand the scope of precision medicine in obstetric care. In this review, we discuss advances, challenges, and possibilities toward the goal of providing proactive, personalized prenatal care. The highlighted advances focus mainly on cell-free nucleic acids; however, we also review research that uses signals from metabolomics, proteomics, intact cells, and the microbiome. We discuss ethical challenges in providing care. Finally, we look to future possibilities, including redefining disease taxonomy and moving from biomarker correlation to biological causation.
Collapse
Affiliation(s)
| | - Diana W Bianchi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development and Section on Prenatal Genomics and Fetal Therapy, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gary M Shaw
- Department of Pediatrics and March of Dimes Prematurity Research Center at Stanford University, Stanford University School of Medicine, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics and March of Dimes Prematurity Research Center at Stanford University, Stanford University School of Medicine, Stanford, California, USA
| | - Stephen R Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, California, USA
- Chan Zuckerberg Initiative, Redwood City, California, USA
| |
Collapse
|
45
|
Costello EK, DiGiulio DB, Robaczewska A, Symul L, Wong RJ, Shaw GM, Stevenson DK, Holmes SP, Kwon DS, Relman DA. Abrupt perturbation and delayed recovery of the vaginal ecosystem following childbirth. Nat Commun 2023; 14:4141. [PMID: 37438386 PMCID: PMC10338445 DOI: 10.1038/s41467-023-39849-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
The vaginal ecosystem is closely tied to human health and reproductive outcomes, yet its dynamics in the wake of childbirth remain poorly characterized. Here, we profile the vaginal microbiota and cytokine milieu of participants sampled longitudinally throughout pregnancy and for at least one year postpartum. We show that delivery, regardless of mode, is associated with a vaginal pro-inflammatory cytokine response and the loss of Lactobacillus dominance. By contrast, neither the progression of gestation nor the approach of labor strongly altered the vaginal ecosystem. At 9.5-months postpartum-the latest timepoint at which cytokines were assessed-elevated inflammation coincided with vaginal bacterial communities that had remained perturbed (highly diverse) from the time of delivery. Time-to-event analysis indicated a one-year postpartum probability of transitioning to Lactobacillus dominance of 49.4%. As diversity and inflammation declined during the postpartum period, dominance by L. crispatus, the quintessential health-associated commensal, failed to return: its prevalence before, immediately after, and one year after delivery was 41%, 4%, and 9%, respectively. Revisiting our pre-delivery data, we found that a prior live birth was associated with a lower odds of L. crispatus dominance in pregnant participants-an outcome modestly tempered by a longer ( > 18-month) interpregnancy interval. Our results suggest that reproductive history and childbirth in particular remodel the vaginal ecosystem and that the timing and degree of recovery from delivery may help determine the subsequent health of the woman and of future pregnancies.
Collapse
Affiliation(s)
- Elizabeth K Costello
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Daniel B DiGiulio
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anna Robaczewska
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Laura Symul
- Department of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Douglas S Kwon
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - David A Relman
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Section of Infectious Diseases, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
46
|
Giles ML, Way SS, Marchant A, Aghaepour N, James T, Schaltz-Buchholzer F, Zazara D, Arck P, Kollmann TR. Maternal Vaccination to Prevent Adverse Pregnancy Outcomes: An Underutilized Molecular Immunological Intervention? J Mol Biol 2023; 435:168097. [PMID: 37080422 PMCID: PMC11533213 DOI: 10.1016/j.jmb.2023.168097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
Adverse pregnancy outcomes including maternal mortality, stillbirth, preterm birth, intrauterine growth restriction cause millions of deaths each year. More effective interventions are urgently needed. Maternal immunization could be one such intervention protecting the mother and newborn from infection through its pathogen-specific effects. However, many adverse pregnancy outcomes are not directly linked to the infectious pathogens targeted by existing maternal vaccines but rather are linked to pathological inflammation unfolding during pregnancy. The underlying pathogenesis driving such unfavourable outcomes have only partially been elucidated but appear to relate to altered immune regulation by innate as well as adaptive immune responses, ultimately leading to aberrant maternal immune activation. Maternal immunization, like all immunization, impacts the immune system beyond pathogen-specific immunity. This raises the possibility that maternal vaccination could potentially be utilised as a pathogen-agnostic immune modulatory intervention to redirect abnormal immune trajectories towards a more favourable phenotype providing pregnancy protection. In this review we describe the epidemiological evidence surrounding this hypothesis, along with the mechanistic plausibility and present a possible path forward to accelerate addressing the urgent need of adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | - Sing Sing Way
- Center for Inflammation and Tolerance; Cincinnati Children's Hospital, Cincinnati, USA
| | | | - Nima Aghaepour
- Stanford University School of Medicine, Stanford, CA, USA
| | - Tomin James
- Stanford University School of Medicine, Stanford, CA, USA
| | | | - Dimitra Zazara
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Petra Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | | |
Collapse
|
47
|
Yüzen D, Graf I, Tallarek AC, Hollwitz B, Wiessner C, Schleussner E, Stammer D, Padula A, Hecher K, Arck PC, Diemert A. Increased late preterm birth risk and altered uterine blood flow upon exposure to heat stress. EBioMedicine 2023:104651. [PMID: 37355458 PMCID: PMC10363435 DOI: 10.1016/j.ebiom.2023.104651] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Climate change, in particular the exposure to heat, impacts on human health and can trigger diseases. Pregnant people are considered a vulnerable group given the physiological changes during pregnancy and the potentially long-lasting consequences for the offspring. Evidence published to date on higher risk of pregnancy complications upon heat stress exposure are from geographical areas with high ambient temperatures. Studies from geographic regions with temperate climates are sparse; however, these areas are critical since individuals may be less equipped to adapt to heat stress. This study addresses a significant gap in knowledge due to the temperature increase documented globally. METHODS Birth data of singleton pregnancies (n = 42,905) from a tertiary care centre in Hamburg, Germany, between 1999 and 2021 were retrospectively obtained and matched with climate data from the warmer season (March to September) provided by the adjacent federal meteorological station of the German National Meteorological Service to calculate the relative risk of heat-associated preterm birth. Heat events were defined by ascending temperature percentiles in combination with humidity over exposure periods of up to 5 days. Further, ultrasound data documented in a longitudinal prospective pregnancy cohort study (n = 612) since 2012 were used to identify pathophysiological causes of heat-induced preterm birth. FINDINGS Both extreme heat and prolonged periods of heat exposure increased the relative risk of preterm birth (RR: 1.59; 95% CI: 1.01-2.43; p = 0.045; RR: 1.20; 95% CI: 1.02-1.40; p = 0.025). We identified a critical period of heat exposure during gestational ages 34-37 weeks that resulted in increased risk of late preterm birth (RR: 1.67; 95% CI: 1.14-1.43; p = 0.009). Pregnancies with a female fetus were more prone to heat stress-associated preterm birth. We found heat exposure was associated with altered vascular resistance within the uterine artery. INTERPRETATION Heat stress caused by high ambient temperatures increases the risk of preterm birth in a geographical region with temperate climate. Prenatal routine care should be revised in such regions to provide active surveillance for women at risk. FUNDING Found in acknowledgements.
Collapse
Affiliation(s)
- Dennis Yüzen
- Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany; Institute of Immunology, University Medical Centre of Hamburg-Eppendorf, Germany
| | - Isabel Graf
- Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany
| | - Ann-Christin Tallarek
- Department of Obstetrics and Fetal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany
| | - Bettina Hollwitz
- Department of Obstetrics and Fetal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany
| | - Christian Wiessner
- Institute of Medical Biometry and Epidemiology, University Medical Centre of Hamburg-Eppendorf, Germany
| | | | - Detlef Stammer
- Centre for Earth System Research and Sustainability (CEN), University Hamburg, Germany
| | - Amy Padula
- Division of Maternal-Fetal Medicine, Department of Obstetrics, University of California, San Francisco, USA
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany
| | - Petra Clara Arck
- Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany.
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Centre of Hamburg-Eppendorf, Germany
| |
Collapse
|
48
|
Yang R, Li X, Ying Z, Zhao Z, Wang Y, Wang Q, Shen B, Peng W. Prematurely delivering mothers show reductions of lachnospiraceae in their gut microbiomes. BMC Microbiol 2023; 23:169. [PMID: 37322412 PMCID: PMC10268532 DOI: 10.1186/s12866-023-02892-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Preterm birth is the leading cause of perinatal morbidity and mortality. Despite evidence shows that imbalances in the maternal microbiome associates to the risk of preterm birth, the mechanisms underlying the association between a perturbed microbiota and preterm birth remain poorly understood. METHOD Applying shotgun metagenomic analysis on 80 gut microbiotas of 43 mothers, we analyzed the taxonomic composition and metabolic function in gut microbial communities between preterm and term mothers. RESULTS Gut microbiome of mothers delivering prematurely showed decreased alpha diversity and underwent significant reorganization, especially during pregnancy. SFCA-producing microbiomes, particularly species of Lachnospiraceae, Ruminococcaceae, and Eubacteriaceae, were significantly depleted in preterm mothers. Lachnospiraceae and its species were the main bacteria contributing to species' differences and metabolic pathways. CONCLUSION Gut microbiome of mothers delivering prematurely has altered and demonstrates the reduction of Lachnospiraceae.
Collapse
Affiliation(s)
- Ru Yang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoyu Li
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Sichuan, China
| | - Zhiye Ying
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Sichuan, China
- Medical Big Data Center, Sichuan University, Chengdu, Sichuan China
| | - Zicheng Zhao
- Shenzhen Byoryn Technology, Shenzhen, Guangdong P.R. China
| | - Yinan Wang
- Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, China
| | - Qingyu Wang
- School of Business Administration, Northeast University, Shenyang, China
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Sichuan, China
| | - Wentao Peng
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
49
|
Babu M, Snyder M. Multi-Omics Profiling for Health. Mol Cell Proteomics 2023; 22:100561. [PMID: 37119971 PMCID: PMC10220275 DOI: 10.1016/j.mcpro.2023.100561] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023] Open
Abstract
The world has witnessed a steady rise in both non-infectious and infectious chronic diseases, prompting a cross-disciplinary approach to understand and treating disease. Current medical care focuses on treating people after they become patients rather than preventing illness, leading to high costs in treating chronic and late-stage diseases. Additionally, a "one-size-fits all" approach to health care does not take into account individual differences in genetics, environment, or lifestyle factors, decreasing the number of people benefiting from interventions. Rapid advances in omics technologies and progress in computational capabilities have led to the development of multi-omics deep phenotyping, which profiles the interaction of multiple levels of biology over time and empowers precision health approaches. This review highlights current and emerging multi-omics modalities for precision health and discusses applications in the following areas: genetic variation, cardio-metabolic diseases, cancer, infectious diseases, organ transplantation, pregnancy, and longevity/aging. We will briefly discuss the potential of multi-omics approaches in disentangling host-microbe and host-environmental interactions. We will touch on emerging areas of electronic health record and clinical imaging integration with muti-omics for precision health. Finally, we will briefly discuss the challenges in the clinical implementation of multi-omics and its future prospects.
Collapse
Affiliation(s)
- Mohan Babu
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
50
|
Espinosa CA, Khan W, Khanam R, Das S, Khalid J, Pervin J, Kasaro MP, Contrepois K, Chang AL, Phongpreecha T, Michael B, Ellenberger M, Mehmood U, Hotwani A, Nizar A, Kabir F, Wong RJ, Becker M, Berson E, Culos A, De Francesco D, Mataraso S, Ravindra N, Thuraiappah M, Xenochristou M, Stelzer IA, Marić I, Dutta A, Raqib R, Ahmed S, Rahman S, Hasan ASMT, Ali SM, Juma MH, Rahman M, Aktar S, Deb S, Price JT, Wise PH, Winn VD, Druzin ML, Gibbs RS, Darmstadt GL, Murray JC, Stringer JSA, Gaudilliere B, Snyder MP, Angst MS, Rahman A, Baqui AH, Jehan F, Nisar MI, Vwalika B, Sazawal S, Shaw GM, Stevenson DK, Aghaeepour N. Multiomic signals associated with maternal epidemiological factors contributing to preterm birth in low- and middle-income countries. SCIENCE ADVANCES 2023; 9:eade7692. [PMID: 37224249 PMCID: PMC10208584 DOI: 10.1126/sciadv.ade7692] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
Preterm birth (PTB) is the leading cause of death in children under five, yet comprehensive studies are hindered by its multiple complex etiologies. Epidemiological associations between PTB and maternal characteristics have been previously described. This work used multiomic profiling and multivariate modeling to investigate the biological signatures of these characteristics. Maternal covariates were collected during pregnancy from 13,841 pregnant women across five sites. Plasma samples from 231 participants were analyzed to generate proteomic, metabolomic, and lipidomic datasets. Machine learning models showed robust performance for the prediction of PTB (AUROC = 0.70), time-to-delivery (r = 0.65), maternal age (r = 0.59), gravidity (r = 0.56), and BMI (r = 0.81). Time-to-delivery biological correlates included fetal-associated proteins (e.g., ALPP, AFP, and PGF) and immune proteins (e.g., PD-L1, CCL28, and LIFR). Maternal age negatively correlated with collagen COL9A1, gravidity with endothelial NOS and inflammatory chemokine CXCL13, and BMI with leptin and structural protein FABP4. These results provide an integrated view of epidemiological factors associated with PTB and identify biological signatures of clinical covariates affecting this disease.
Collapse
Affiliation(s)
- Camilo A. Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sayan Das
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Javairia Khalid
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Margaret P. Kasaro
- University of North Carolina Global Projects Zambia, Lusaka, Zambia
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanaphong Phongpreecha
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ambreen Nizar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Neal Ravindra
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Melan Thuraiappah
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Arup Dutta
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Rubhana Raqib
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | | | | | - Said M. Ali
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Mohamed H. Juma
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Monjur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Shaki Aktar
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Saikat Deb
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Joan T. Price
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Paul H. Wise
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurice L. Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald S. Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary L. Darmstadt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeffrey S. A. Stringer
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anisur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Abdullah H. Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fyezah Jehan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Bellington Vwalika
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Sunil Sazawal
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|