Bio


Lawrence (Lance) S. Prince, MD, PhD, is the Division Chief for Neonatal and Developmental Medicine at Stanford School of Medicine. Dr. Prince was previously a Professor of Pediatrics and Chief of the Division of Neonatology at the University of California, San Diego and Rady Children’s Hospital, San Diego.

Dr. Prince has a long and distinguished career mentoring clinical and scientific trainees and students, many of whom have gone on to establish their own successful careers as academic physician investigators. As a physician scientist, Dr. Prince leads a basic science laboratory focusing on the mechanisms regulating developmental immunology and lung injury and repair. Dr. Prince received a Bachelor of Science in Chemistry from University of Miami, an MD/PhD with a focus in Cell Biology from University of Alabama at Birmingham, and postdoctoral fellowship, Pediatrics residency, and Neonatal-Perinatal Medicine Fellowship training at the University of Iowa. Before arriving in California, Dr. Prince was an Associate Professor of Pediatrics at Vanderbilt University.

Dr. Prince’s research interests include the molecular and cellular mechanisms controlling lung development and the maturation of the fetal and neonatal immune systems. He has a particular clinical interest in managing and treating neonatal lung diseases, especially bronchopulmonary dysplasia (BPD) in babies born extremely preterm. Dr. Prince’s research team focuses primarily on the development of innate immunity during fetal life as it impacts health and disease in preterm infants. The laboratory is investigating how microbes including Group B streptococcus exploit the unique features of neonatal macrophages to avoid immune detection and cause disease, as well as leading a number of clinical and translational investigations.

Clinical Focus


  • Neonatal-Perinatal Medicine

Academic Appointments


Administrative Appointments


  • Chief, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine (2020 - Present)
  • Professor of Pediatrics, Stanford University School of Medicine (2020 - Present)
  • Co-Director, Johnson Center for Pregnancy and Newborn Services, Lucile Packard Children's Hospital (2020 - Present)

Honors & Awards


  • Fellow Basic Research Award, Society for Pediatric Research (1999)
  • Trainee Investigator Award, Midwest Society for Pediatric Research (2000)
  • National Staff Research Award, American Lung Association (2005)
  • Philip Sunshine, M.D Endowed Professorship in Neonatology, Stanford University School of Medicine (2020)

Boards, Advisory Committees, Professional Organizations


  • Member, Society for Pediatric Research (2007 - Present)
  • Member, American Association of Immunologists (2011 - Present)
  • Member, Western Society for Pediatric Research (2014 - Present)
  • Member, American Pediatric Society (2023 - Present)
  • Member, Lucile Packard Children's Hospital Medical Staff Executive Committee (2024 - Present)

Professional Education


  • Fellowship: University of Iowa Hospitals and Clinics (2002) IA
  • Residency: University of Iowa Hospitals and Clinics (2000) IA
  • Medical Education: University of Alabama at Birmingham School of Medicine (1996) AL
  • Board Certification: American Board of Pediatrics, Neonatal-Perinatal Medicine (2005)

Stanford Advisees


All Publications


  • Macrophage Polarizations in the Placenta and Lung are Associated with Bronchopulmonary Dysplasia. bioRxiv : the preprint server for biology Mestan, K. K., Sharma, A., Lazar, S., Pandey, S., Parast, M. M., Laurent, L. C., Prince, L. S., Sahoo, D. 2024

    Abstract

    The intricate interplay between macrophage polarization and placenta vascular dysfunction has garnered increasing attention in the context of placental inflammatory diseases. This study delves into the complex relationship between macrophage polarization within the placenta and its potential impact on the development of vascular dysfunction and inflammatory conditions. The placenta, a crucial organ in fetal development, relies on a finely tuned balance of immune responses for proper functioning. Disruptions in this delicate equilibrium can lead to pathological conditions, including inflammatory diseases affecting the fetus and newborn infant. We explored the interconnectedness between placental macrophage polarization and its relevance to lung macrophages, particularly in the context of early life lung development. Bronchopulmonary dysplasia (BPD), the most common chronic lung disease of prematurity, has been associated with abnormal immune responses, and understanding the role of macrophages in this context is pivotal. The investigation aims to shed light on how alterations in placental macrophage polarization may contribute to lung macrophage behavior and, consequently, influence the development of BPD. By unraveling the intricate mechanisms linking macrophage polarization, placental dysfunction and BPD, this research seeks to provide insights that could pave the way for targeted therapeutic interventions. The findings may offer novel perspectives on preventing and managing placental and lung-related pathologies, ultimately contributing to improved maternal and neonatal health outcomes.

    View details for DOI 10.1101/2024.01.26.577443

    View details for PubMedID 38352616

    View details for PubMedCentralID PMC10862768

  • Advances and potential of omics studies for understanding the development of food allergy. Frontiers in allergy Sindher, S. B., Chin, A. R., Aghaeepour, N., Prince, L., Maecker, H., Shaw, G. M., Stevenson, D. K., Nadeau, K. C., Snyder, M., Khatri, P., Boyd, S. D., Winn, V. D., Angst, M. S., Chinthrajah, R. S. 2023; 4: 1149008

    Abstract

    The prevalence of food allergy continues to rise globally, carrying with it substantial safety, economic, and emotional burdens. Although preventative strategies do exist, the heterogeneity of allergy trajectories and clinical phenotypes has made it difficult to identify patients who would benefit from these strategies. Therefore, further studies investigating the molecular mechanisms that differentiate these trajectories are needed. Large-scale omics studies have identified key insights into the molecular mechanisms for many different diseases, however the application of these technologies to uncover the drivers of food allergy development is in its infancy. Here we review the use of omics approaches in food allergy and highlight key gaps in knowledge for applying these technologies for the characterization of food allergy development.

    View details for DOI 10.3389/falgy.2023.1149008

    View details for PubMedID 37034151

    View details for PubMedCentralID PMC10080041

  • Data-driven longitudinal characterization of neonatal health and morbidity. Science translational medicine De Francesco, D., Reiss, J. D., Roger, J., Tang, A. S., Chang, A. L., Becker, M., Phongpreecha, T., Espinosa, C., Morin, S., Berson, E., Thuraiappah, M., Le, B. L., Ravindra, N. G., Payrovnaziri, S. N., Mataraso, S., Kim, Y., Xue, L., Rosenstein, M. G., Oskotsky, T., Marić, I., Gaudilliere, B., Carvalho, B., Bateman, B. T., Angst, M. S., Prince, L. S., Blumenfeld, Y. J., Benitz, W. E., Fuerch, J. H., Shaw, G. M., Sylvester, K. G., Stevenson, D. K., Sirota, M., Aghaeepour, N. 2023; 15 (683): eadc9854

    Abstract

    Although prematurity is the single largest cause of death in children under 5 years of age, the current definition of prematurity, based on gestational age, lacks the precision needed for guiding care decisions. Here, we propose a longitudinal risk assessment for adverse neonatal outcomes in newborns based on a deep learning model that uses electronic health records (EHRs) to predict a wide range of outcomes over a period starting shortly before conception and ending months after birth. By linking the EHRs of the Lucile Packard Children's Hospital and the Stanford Healthcare Adult Hospital, we developed a cohort of 22,104 mother-newborn dyads delivered between 2014 and 2018. Maternal and newborn EHRs were extracted and used to train a multi-input multitask deep learning model, featuring a long short-term memory neural network, to predict 24 different neonatal outcomes. An additional cohort of 10,250 mother-newborn dyads delivered at the same Stanford Hospitals from 2019 to September 2020 was used to validate the model. Areas under the receiver operating characteristic curve at delivery exceeded 0.9 for 10 of the 24 neonatal outcomes considered and were between 0.8 and 0.9 for 7 additional outcomes. Moreover, comprehensive association analysis identified multiple known associations between various maternal and neonatal features and specific neonatal outcomes. This study used linked EHRs from more than 30,000 mother-newborn dyads and would serve as a resource for the investigation and prediction of neonatal outcomes. An interactive website is available for independent investigators to leverage this unique dataset: https://maternal-child-health-associations.shinyapps.io/shiny_app/.

    View details for DOI 10.1126/scitranslmed.adc9854

    View details for PubMedID 36791208

  • Sp3 is essential for normal lung morphogenesis and cell cycle progression during mouse embryonic development. Development (Cambridge, England) McCoy, A. M., Lakhdari, O., Shome, S., Caoili, K., Hernandez, G. E., Aghaeepour, N., Butcher, L. D., Fisch, K., Prince, L. S. 2023

    Abstract

    Members of the Sp family of transcription factors regulate gene expression via binding GC boxes within promoter regions. Unlike Sp1, which stimulates transcription, the closely related Sp3 can either repress or activate gene expression and is required for perinatal survival in mice. Here we use RNAseq and cellular phenotyping to show how Sp3 regulates murine fetal cell differentiation and proliferation. Homozygous Sp3-/- mice were smaller than WT and Sp+/- littermates, died soon after birth, and had abnormal lung morphogenesis. RNAseq of Sp3-/- fetal lung mesenchymal cells identified alterations in extracellular matrix production, developmental signaling pathways, and myofibroblast/lipofibroblast differentiation. The lungs of Sp3-/- mice contained multiple structural defects, with abnormal endothelial cell morphology, lack of elastic fiber formation, and accumulation of lipid droplets within mesenchymal lipofibroblasts. Sp3-/- cells and mice also displayed cell cycle arrest, with accumulation in G0/G1 and reduced expression of numerous cell cycle regulators including Ccne1. These data detail the global impact of Sp3 on in vivo mouse gene expression and development.

    View details for DOI 10.1242/dev.200839

    View details for PubMedID 36762637

  • Progressive Metabolic Abnormalities Associated with the Development of Neonatal Bronchopulmonary Dysplasia. Nutrients Ye, C., Wu, J., Reiss, J. D., Sinclair, T. J., Stevenson, D. K., Shaw, G. M., Chace, D. H., Clark, R. H., Prince, L. S., Ling, X. B., Sylvester, K. G. 2022; 14 (17)

    Abstract

    Objective: To assess the longitudinal metabolic patterns during the evolution of bronchopulmonary dysplasia (BPD) development. Methods: A case-control dataset of preterm infants (<32-week gestation) was obtained from a multicenter database, including 355 BPD cases and 395 controls. A total of 72 amino acid (AA) and acylcarnitine (AC) variables, along with infants' calorie intake and growth outcomes, were measured on day of life 1, 7, 28, and 42. Logistic regression, clustering methods, and random forest statistical modeling were utilized to identify metabolic variables significantly associated with BPD development and to investigate their longitudinal patterns that are associated with BPD development. Results: A panel of 27 metabolic variables were observed to be longitudinally associated with BPD development. The involved metabolites increased from 1 predominant different AC by day 7 to 19 associated AA and AC compounds by day 28 and 16 metabolic features by day 42. Citrulline, alanine, glutamate, tyrosine, propionylcarnitine, free carnitine, acetylcarnitine, hydroxybutyrylcarnitine, and most median-chain ACs (C5:C10) were the most associated metabolites down-regulated in BPD babies over the early days of life, whereas phenylalanine, methionine, and hydroxypalmitoylcarnitine were observed to be up-regulated in BPD babies. Most calorie intake and growth outcomes revealed similar longitudinal patterns between BPD cases and controls over the first 6 weeks of life, after gestational adjustment. When combining with birth weight, the derived metabolic-based discriminative model observed some differences between those with and without BPD development, with c-statistics of 0.869 and 0.841 at day 7 and 28 of life on the test data. Conclusions: The metabolic panel we describe identified some metabolic differences in the blood associated with BPD pathogenesis. Further work is needed to determine whether these compounds could facilitate the monitoring and/or investigation of early-life metabolic status in the lung and other tissues for the prevention and management of BPD.

    View details for DOI 10.3390/nu14173547

    View details for PubMedID 36079804

  • The Lung Microenvironment Instructs Gene Transcription in Neonatal and Adult Alveolar Macrophages. Journal of immunology (Baltimore, Md. : 1950) Honda, A., Hoeksema, M. A., Sakai, M., Lund, S. J., Lakhdari, O., Butcher, L. D., Rambaldo, T. C., Sekiya, N. M., Nasamran, C. A., Fisch, K. M., Sajti, E., Glass, C. K., Prince, L. S. 2022

    Abstract

    Immaturity of alveolar macrophages (AMs) around birth contributes to the susceptibility of newborns to lung disease. However, the molecular features differentiating neonatal and mature, adult AMs are poorly understood. In this study, we identify the unique transcriptomes and enhancer landscapes of neonatal and adult AMs in mice. Although the core AM signature was similar, murine adult AMs expressed higher levels of genes involved in lipid metabolism, whereas neonatal AMs expressed a largely proinflammatory gene profile. Open enhancer regions identified by an assay for transposase-accessible chromatin followed by high-throughput sequencing (ATAC-seq) contained motifs for nuclear receptors, MITF, and STAT in adult AMs and AP-1 and NF-kappaB in neonatal AMs. Intranasal LPS activated a similar innate immune response in both neonatal and adult mice, with higher basal expression of inflammatory genes in neonates. The lung microenvironment drove many of the distinguishing gene expression and open chromatin characteristics of neonatal and adult AMs. Neonatal mouse AMs retained high expression of some proinflammatory genes, suggesting that the differences in neonatal AMs result from both inherent cell properties and environmental influences.

    View details for DOI 10.4049/jimmunol.2101192

    View details for PubMedID 35354612

  • Antenatal Mesenchymal Stromal Cell Extracellular Vesicle Therapy Prevents Preeclamptic Lung Injury in Mice. American journal of respiratory cell and molecular biology Taglauer, E. S., Fernandez-Gonzalez, A., Willis, G. R., Reis, M., Yeung, V., Liu, X., Prince, L. S., Mitsialis, S. A., Kourembanas, S. 2021

    Abstract

    In preeclamptic pregnancies, a variety of intrauterine alterations lead to abnormal placentation, release of inflammatory/antiangiogenic factors, and subsequent fetal growth restriction with significant potential to cause a primary insult to the developing fetal lung. Thus, modulation of the maternal intrauterine environment may be a key therapeutic avenue to prevent preeclampsia-associated developmental lung injury. A biologic therapy of interest are mesenchymal stromal cell-derived extracellular vesicles (MEx), which we have previously shown to ameliorate preeclamptic physiology through intrauterine immunomodulation. To evaluate the therapeutic potential of MEx to improve developmental lung injury in experimental preeclampsia. Using the heme oxygenase-1 null mouse (Hmox1-/-) model, preeclamptic pregnant dams were administered intravenous antenatal MEx treatment during each week of pregnancy followed by analysis of fetal and postnatal lung tissues, amniotic fluid protein profiles and lung explant/amniotic fluid co-cultures in comparison with control and untreated preeclamptic pregnancies. We first identified that a preeclamptic intrauterine environment had a significant adverse impact on fetal lung development including alterations in fetal lung developmental gene profiles in addition to postnatal alveolar and bronchial changes. Amniotic fluid proteomic analysis and fetal lung explant/amniotic fluid co-cultures further demonstrated that maternally administered MEx altered the expression of multiple inflammatory mediators in the preeclamptic intrauterine compartment resulting in normalization of fetal lung branching morphogenesis and developmental gene expression. Our evaluation of fetal and postnatal parameters overall suggests that antenatal MEx treatment may provide a highly valuable preventative therapeutic modality for amelioration of lung development in preeclamptic disease.

    View details for DOI 10.1165/rcmb.2021-0307OC

    View details for PubMedID 34614384

  • Black swans and ambitious overgeneralization in newborn intensive care. Pediatric research Stevenson, D. K., Wong, R. J., Shaw, G. M., Aghaeepour, N., Maric, I., Prince, L. S., Reiss, J. D., Katz, M. 2021

    View details for DOI 10.1038/s41390-021-01771-5

    View details for PubMedID 34601493

  • 50 Years Ago in TheJournalofPediatrics: Neonatal Hypoglycemia: Progress and Predicaments. The Journal of pediatrics Ikle, J. M., Prince, L. S., Maahs, D. M. 2021; 235: 82

    View details for DOI 10.1016/j.jpeds.2021.05.001

    View details for PubMedID 34304767

  • An RCT of Rapid Genomic Sequencing among Seriously Ill Infants Results in High Clinical Utility, Changes in Management, and Low Perceived Harm. American journal of human genetics Dimmock, D. P., Clark, M. M., Gaughran, M., Cakici, J. A., Caylor, S. A., Clarke, C., Feddock, M., Chowdhury, S., Salz, L., Cheung, C., Bird, L. M., Hobbs, C., Wigby, K., Farnaes, L., Bloss, C. S., Kingsmore, S. F., RCIGM Investigators, Bainbridge, M. N., Barea, J., Batalov, S., Bezares, Z., Bird, L. M., Bloss, C. S., Braun, J. J., Cakici, J. A., Del Campo, M., Carroll, J., Cheung, C., Cohenmeyer, C., Coufal, N. G., Diaz, C., Ding, Y., Ellsworth, K., Evans, M., Feigenbaum, A., Friedman, J., Gleeson, J., Hansen, C., Honold, J., James, K., Jones, M. C., Kimball, A., Knight, G., Van Der Kraan, L., Lane, B., Le, J., Leibel, S., Lenberg, J., Mashburn, D., Moyer, L., Mulrooney, P., Nahas, S., Oh, D., Orendain, D., Oriol, A., Ortiz-Arechiga, M., Prince, L., Rego, S., Reyes, I., Sanford, E., Sauer, C., Schwanemann, L., Speziale, M., Suttner, D., Sweeney, N., Song, R., Tokita, M., Veeraraghavan, N., Watkins, K., Wong, T., Wright, M. S., Yamada, C. 2020; 107 (5): 942–52

    Abstract

    The second Newborn Sequencing in Genomic Medicine and Public Health (NSIGHT2) study was a randomized, controlled trial of rapid whole-genome sequencing (rWGS) or rapid whole-exome sequencing (rWES) in infants with diseases of unknown etiology in intensive care units (ICUs). Gravely ill infants were not randomized and received ultra-rapid whole-genome sequencing (urWGS). Herein we report results of clinician surveys of the clinical utility of rapid genomic sequencing (RGS). The primary end-point-clinician perception that RGS was useful- was met for 154 (77%) of 201 infants. Both positive and negative tests were rated as having clinical utility (42 of 45 [93%] and 112 of 156 [72%], respectively). Physicians reported that RGS changed clinical management in 57 (28%) infants, particularly in those receiving urWGS (p = 0.0001) and positive tests (p < 0.00001). Outcomes of 32 (15%) infants were perceived to be changed by RGS. Positive tests changed outcomes more frequently than negative tests (p < 0.00001). In logistic regression models, the likelihood that RGS was perceived as useful increased 6.7-fold when associated with changes in management (95% CI 1.8-43.3). Changes in management were 10.1-fold more likely when results were positive (95% CI 4.7-22.4) and turnaround time was shorter (odds ratio 0.92, 95% CI 0.85-0.99). RGS seldom led to clinician-perceived confusion or distress among families (6 of 207 [3%]). In summary, clinicians perceived high clinical utility and low likelihood of harm with first-tier RGS of infants in ICUs with diseases of unknown etiology. RGS was perceived as beneficial irrespective of whether results were positive or negative.

    View details for DOI 10.1016/j.ajhg.2020.10.003

    View details for PubMedID 33157007

  • Developmental Immaturity of Siglec Receptor Expression on Neonatal Alveolar Macrophages Predisposes to Severe Group B Streptococcal Infection ISCIENCE Lund, S. J., Patras, K. A., Kimmey, J. M., Yamamura, A., Butcher, L. D., Del Rosario, P. B., Hernandez, G. E., McCoy, A. M., Lakhdari, O., Nizet, V., Prince, L. S. 2020; 23 (6): 101207

    Abstract

    Streptococcus agalactiae (Group B Streptococcus, GBS) is the most common neonatal pathogen. However, the cellular and molecular mechanisms for neonatal susceptibility to GBS pneumonia and sepsis are incompletely understood. Here we optimized a mouse model of GBS pneumonia to test the role of alveolar macrophage (ΑΜΦ) maturation in host vulnerability to disease. Compared with juvenile and adult mice, neonatal mice infected with GBS had increased mortality and persistence of lung injury. In addition, neonatal mice were defective in GBS phagocytosis and killing. ΑΜΦ depletion and disruption of ΑΜΦ differentiation in Csf2-/- mice both impaired GBS clearance. AMΦ engage the heavily sialylated GBS capsule via the cell surface Siglec receptors Sn and Siglec-E. Although both newborn and adult ΑΜΦ expressed Siglec-E, newborn ΑΜΦ expressed significantly lower levels of Sn. We propose that a developmental delay in Sn expression on ΑΜΦ may prevent effective killing and clearing of GBS from the newborn lung.

    View details for DOI 10.1016/j.isci.2020.101207

    View details for Web of Science ID 000548240300006

    View details for PubMedID 32535023

    View details for PubMedCentralID PMC7300150

  • Transcriptional profiling of lung macrophages identifies a predictive signature for inflammatory lung disease in preterm infants COMMUNICATIONS BIOLOGY Sahoo, D., Zaramela, L. S., Hernandez, G. E., Mai, U., Taheri, S., Dang, D., Stouch, A. N., Medal, R. M., McCoy, A. M., Aschner, J. L., Blackwell, T. S., Zengler, K., Prince, L. S. 2020; 3 (1): 259

    Abstract

    Lung macrophages mature after birth, placing newborn infants, particularly those born preterm, within a unique window of susceptibility to disease. We hypothesized that in preterm infants, lung macrophage immaturity contributes to the development of bronchopulmonary dysplasia (BPD), the most common serious complication of prematurity. By measuring changes in lung macrophage gene expression in preterm patients at risk of BPD, we show here that patients eventually developing BPD had higher inflammatory mediator expression even on the first day of life. Surprisingly, the ex vivo response to LPS was similar across all samples. Our analysis did however uncover macrophage signature genes whose expression increased in the first week of life specifically in patients resilient to disease. We propose that these changes describe the dynamics of human lung macrophage differentiation. Our study therefore provides new mechanistic insight into both neonatal lung disease and human developmental immunology.

    View details for DOI 10.1038/s42003-020-0985-2

    View details for Web of Science ID 000537108300008

    View details for PubMedID 32444859

    View details for PubMedCentralID PMC7244484

  • Computational Approach to Identifying Universal Macrophage Biomarkers FRONTIERS IN PHYSIOLOGY Dang, D., Taheri, S., Das, S., Ghosh, P., Prince, L. S., Sahoo, D. 2020; 11: 275

    Abstract

    Macrophages engulf and digest microbes, cellular debris, and various disease-associated cells throughout the body. Understanding the dynamics of macrophage gene expression is crucial for studying human diseases. As both bulk RNAseq and single cell RNAseq datasets become more numerous and complex, identifying a universal and reliable marker of macrophage cell becomes paramount. Traditional approaches have relied upon tissue specific expression patterns. To identify universal biomarkers of macrophage, we used a previously published computational approach called BECC (Boolean Equivalent Correlated Clusters) that was originally used to identify conserved cell cycle genes. We performed BECC analysis using the known macrophage marker CD14 as a seed gene. The main idea behind BECC is that it uses massive database of public gene expression dataset to establish robust co-expression patterns identified using a combination of correlation, linear regression and Boolean equivalences. Our analysis identified and validated FCER1G and TYROBP as novel universal biomarkers for macrophages in human and mouse tissues.

    View details for DOI 10.3389/fphys.2020.00275

    View details for Web of Science ID 000529017900001

    View details for PubMedID 32322218

    View details for PubMedCentralID PMC7156600

  • Transcriptomic and epigenetic mechanisms underlying myeloid diversity in the lung NATURE IMMUNOLOGY Sajti, E., Link, V. M., Ouyang, Z., Spann, N. J., Westin, E., Romanoski, C. E., Fonseca, G. J., Prince, L. S., Glass, C. K. 2020; 21 (2): 221-+

    Abstract

    The lung is inhabited by resident alveolar and interstitial macrophages as well as monocytic cells that survey lung tissues. Each cell type plays distinct functional roles under homeostatic and inflammatory conditions, but mechanisms establishing their molecular identities and functional potential remain poorly understood. In the present study, systematic evaluation of transcriptomes and open chromatin of alveolar macrophages (AMs), interstitial macrophages (IMs) and lung monocytes from two mouse strains enabled inference of common and cell-specific transcriptional regulators. We provide evidence that these factors drive selection of regulatory landscapes that specify distinct phenotypes of AMs and IMs and entrain qualitatively different responses to toll-like receptor 4 signaling in vivo. These studies reveal a striking divergence in a fundamental innate immune response pathway in AMs and establish a framework for further understanding macrophage diversity in the lung.

    View details for DOI 10.1038/s41590-019-0582-z

    View details for Web of Science ID 000508964100001

    View details for PubMedID 31959980

    View details for PubMedCentralID PMC7667722

  • A Randomized, Controlled Trial of the Analytic and Diagnostic Performance of Singleton and Trio, Rapid Genome and Exome Sequencing in III Infants AMERICAN JOURNAL OF HUMAN GENETICS Kingsmore, S. F., Cakici, J. A., Clark, M. M., Gaughran, M., Feddock, M., Batalov, S., Bainbridge, M. N., Carroll, J., Caylor, S. A., Clarke, C., Ding, Y., Ellsworth, K., Farnaes, L., Hildreth, A., Hobbs, C., James, K., Kint, C. I., Lenberg, J., Nahas, S., Prince, L., Reyes, I., Salz, L., Sanford, E., Schols, P., Sweeney, N., Tokita, M., Veeraraghavan, N., Watkins, K., Wigby, K., Wong, T., Chowdhury, S., Wright, M. S., Dimmock, D., Bezares, Z., Bloss, C., Braun, J. A., Diaz, C., Mashburn, D., Tamang, D., Orendain, D., Friedman, J., Gleeson, J., Barea, J., Chiang, G., Cohenmeyer, C., Coufal, N. G., Evans, M., Honold, J., Hovey, R. L., Kimball, A., Lane, B., Le, C., Le, J., Leibel, S., Moyer, L., Mulrooney, P., Oh, D., Ordonez, P., Oriol, A., Ortiz-Arechiga, M., Puckett, L., Speziale, M., Suttner, D., Van der Kraan, L., Knight, G., Sauer, C., Song, R., White, S., Wise, A., Yamada, C., RCIGM Investigators 2019; 105 (4): 719–33

    Abstract

    The second Newborn Sequencing in Genomic Medicine and Public Health study was a randomized, controlled trial of the effectiveness of rapid whole-genome or -exome sequencing (rWGS or rWES, respectively) in seriously ill infants with diseases of unknown etiology. Here we report comparisons of analytic and diagnostic performance. Of 1,248 ill inpatient infants, 578 (46%) had diseases of unknown etiology. 213 infants (37% of those eligible) were enrolled within 96 h of admission. 24 infants (11%) were very ill and received ultra-rapid whole-genome sequencing (urWGS). The remaining infants were randomized, 95 to rWES and 94 to rWGS. The analytic performance of rWGS was superior to rWES, including variants likely to affect protein function, and ClinVar pathogenic/likely pathogenic variants (p < 0.0001). The diagnostic performance of rWGS and rWES were similar (18 diagnoses in 94 infants [19%] versus 19 diagnoses in 95 infants [20%], respectively), as was time to result (median 11.0 versus 11.2 days, respectively). However, the proportion diagnosed by urWGS (11 of 24 [46%]) was higher than rWES/rWGS (p = 0.004) and time to result was less (median 4.6 days, p < 0.0001). The incremental diagnostic yield of reflexing to trio after negative proband analysis was 0.7% (1 of 147). In conclusion, rapid genomic sequencing can be performed as a first-tier diagnostic test in inpatient infants. urWGS had the shortest time to result, which was important in unstable infants, and those in whom a genetic diagnosis was likely to impact immediate management. Further comparison of urWGS and rWES is warranted because genomic technologies and knowledge of variant pathogenicity are evolving rapidly.

    View details for DOI 10.1016/j.ajhg.2019.08.009

    View details for Web of Science ID 000488953400004

    View details for PubMedID 31564432

    View details for PubMedCentralID PMC6817534

  • Differential Immune Activation in Fetal Macrophage Populations SCIENTIFIC REPORTS Lakhdari, O., AsamiYamamura, Hernandez, G. E., Anderson, K. K., Lund, S. J., Oppong-Nonterah, G. O., Hoffman, H. M., Prince, L. S. 2019; 9: 7677

    Abstract

    Distinct macrophage subsets populate the developing embryo and fetus in distinct waves. However little is known about the functional differences between in utero macrophage populations or how they might contribute to fetal and neonatal immunity. Here we tested the innate immune response of mouse macrophages derived from the embryonic yolk sac and from fetal liver. When isolated from liver or lung, CD11bHI fetal liver derived macrophages responded to the TLR4 agonist LPS by expressing and releasing inflammatory cytokines. However F4/80HI macrophages from the yolk sac did not respond to LPS treatment. While differences in TLR4 expression did not appear to explain these data, F4/80HI macrophages had much lower NLRP3 inflammasome expression compared to CD11bHI macrophages. Gene expression profiling also demonstrated LPS-induced expression of inflammatory genes in CD11bHI macrophages, but not in F4/80HI cells. Genes expressed in LPS-treated CD11bHI macrophages were more likely to contain predicted NF-κB binding sites in their promoter regions. Our data show that CD11bHI macrophages derived from fetal liver are the major pro-inflammatory cells in the developing fetus. These findings could have important implications in better understanding the fetal inflammatory response and the unique features of neonatal immunity.

    View details for DOI 10.1038/s41598-019-44181-8

    View details for Web of Science ID 000468600100003

    View details for PubMedID 31118442

    View details for PubMedCentralID PMC6531440

  • TLR Activation Alters Bone Marrow-Derived Macrophage Differentiation JOURNAL OF INNATE IMMUNITY Oppong-Nonterah, G. O., Lakhdari, O., Yamamura, A., Hoffman, H. M., Prince, L. S. 2019; 11 (1): 99–108

    Abstract

    Early exposure to inflammatory signals may have a lasting impact on immune function. Present throughout embryogenesis, macrophages are key cells providing innate immune protection to the developing fetus and newborn. Here, we have used an established model of macrophage development to test how early inflammatory signals can impact cellular differentiation and function. Bone marrow-derived macrophages were treated with Escherichia coli lipopolysaccharide (LPS) 2 days after initial isolation and culture. LPS treatment during this early stage of differentiation decreased the expression of CSF1R and increased that of the mature macrophage marker F4/80. These early changes in macrophage differentiation were also measured in cells from mice lacking IKKβ, but the change in CSF1R expression after LPS treatment was blocked with MAPK inhibition. LPS-induced changes in macrophage marker expression persisted following LPS removal, suggesting that early inflammatory activation could induce a lasting developmental impact. Early LPS exposure inhibited macrophage phagocytosis of labeled E. coli while LPS had no effect on fully differentiated macrophages. Our data demonstrate that early inflammatory exposure to a microbial stimulus induce lasting phenotypic changes in macrophages.

    View details for DOI 10.1159/000494070

    View details for Web of Science ID 000455065600010

    View details for PubMedID 30408777

    View details for PubMedCentralID PMC6296861

  • FGF10 and Human Lung Disease Across the Life Spectrum FRONTIERS IN GENETICS Prince, L. S. 2018; 9: 517

    Abstract

    Lung diseases impact patients across the lifespan, from infants in the first minutes of life through the aged population. Congenital abnormalities of lung structure can cause lung disease at birth or make adults more susceptible to chronic disease. Continuous inhalation of atmospheric components also requires the lung to be resilient to cellular injury. Fibroblast growth factor 10 (FGF10) regulates multiple stages of structural lung morphogenesis, cellular differentiation, and the response to injury. As a driver of lung airway branching morphogenesis, FGF10 signaling defects during development lead to neonatal lung disease. Alternatively, congenital airway abnormalities attributed to FGF10 mutations increase the risk of chronic airway disease in adulthood. FGF10 also maintains progenitor cell populations in the airway and promotes alveolar type 2 cell expansion and differentiation following injury. Here we review the cellular and molecular mechanisms linking FGF10 to multiple lung diseases, from bronchopulmonary dysplasia in extremely preterm neonates, cystic fibrosis in children, and chronic adult lung disorders. Understanding the connections between FGF10 and lung diseases may lead to exciting new therapeutic strategies.

    View details for DOI 10.3389/fgene.2018.00517

    View details for Web of Science ID 000448908900001

    View details for PubMedID 30429870

    View details for PubMedCentralID PMC6220039

  • IKK beta Activation in the Fetal Lung Mesenchyme Alters Lung Vascular Development but Not Airway Morphogenesis AMERICAN JOURNAL OF PATHOLOGY McCoy, A. M., Herington, J. L., Stouch, A. N., Mukherjee, A. B., Lakhdari, O., Blackwel, T. S., Prince, L. S. 2017; 187 (12): 2635–44

    Abstract

    In the immature lung, inflammation and injury disrupt the epithelial-mesenchymal interactions required for normal development. Innate immune signaling and NF-κB activation disrupt the normal expression of multiple mesenchymal genes that play a key role in airway branching and alveolar formation. To test the role of the NF-κB pathway specifically in lung mesenchyme, we utilized the mesenchymal Twist2-Cre to drive expression of a constitutively active inhibitor of NF-κB kinase subunit β (IKKβca) mutant in developing mice. Embryonic Twist2-IKKβca mice were generated in expected numbers and appeared grossly normal. Airway branching also appeared normal in Twist2-IKKβca embryos, with airway morphometry, elastin staining, and saccular branching similar to those in control littermates. While Twist2-IKKβca lungs did not contain increased levels of Il1b, we did measure an increased expression of the chemokine-encoding gene Ccl2. Twist2-IKKβca lungs had increased staining for the vascular marker platelet endothelial cell adhesion molecule 1. In addition, type I alveolar epithelial differentiation appeared to be diminished in Twist2-IKKβca lungs. The normal airway branching and lack of Il1b expression may have been due to the inability of the Twist2-IKKβca transgene to induce inflammasome activity. While Twist2-IKKβca lungs had an increased number of macrophages, inflammasome expression remained restricted to macrophages without evidence of spontaneous inflammasome activity. These results emphasize the importance of cellular niche in considering how inflammatory signaling influences fetal lung development.

    View details for DOI 10.1016/j.ajpath.2017.08.013

    View details for Web of Science ID 000417009100003

    View details for PubMedID 28923684

    View details for PubMedCentralID PMC5718091

  • The innate immune response in fetal lung mesenchymal cells targets VEGFR2 expression and activity AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY Medal, R. M., Im, A. M., Yamamoto, Y., Lakhdari, O., Blackwell, T. S., Hoffman, H. M., Sahoo, D., Prince, L. S. 2017; 312 (6): L861–L872

    Abstract

    In preterm infants, soluble inflammatory mediators target lung mesenchymal cells, disrupting airway and alveolar morphogenesis. However, how mesenchymal cells respond directly to microbial stimuli remains poorly characterized. Our objective was to measure the genome-wide innate immune response in fetal lung mesenchymal cells exposed to the bacterial endotoxin lipopolysaccharide (LPS). With the use of Affymetrix MoGene 1.0st arrays, we showed that LPS induced expression of unique innate immune transcripts heavily weighted toward CC and CXC family chemokines. The transcriptional response was different between cells from E11, E15, and E18 mouse lungs. In all cells tested, LPS inhibited expression of a small core group of genes including the VEGF receptor Vegfr2 Although best characterized in vascular endothelial populations, we demonstrated here that fetal mouse lung mesenchymal cells express Vegfr2 and respond to VEGF-A stimulation. In mesenchymal cells, VEGF-A increased cell migration, activated the ERK/AKT pathway, and promoted FOXO3A nuclear exclusion. With the use of an experimental coculture model of epithelial-mesenchymal interactions, we also showed that VEGFR2 inhibition prevented formation of three-dimensional structures. Both LPS and tyrosine kinase inhibition reduced three-dimensional structure formation. Our data suggest a novel mechanism for inflammation-mediated defects in lung development involving reduced VEGF signaling in lung mesenchyme.

    View details for DOI 10.1152/ajplung.00554.2016

    View details for Web of Science ID 000404384100008

    View details for PubMedID 28336813

    View details for PubMedCentralID PMC5495951

  • TLR3 signaling is downregulated by a MAVS isoform in epithelial cells CELLULAR IMMUNOLOGY Lakhdari, O., McAllister, C. S., Wang, M., Minev, I., Prince, L. S., Eckmann, L., Kagnoff, M. F. 2016; 310: 205–10

    Abstract

    Innate immune responses to dsRNA result in signaling through the TLR3 pathway and/or the RIG-I/MDA-5/MAVS pathway which can activate type I IFN, proinflammatory cytokines and apoptosis. It is not clear whether MAVS could play a role in TLR3-dependent responses to extracellular dsRNA. Using a model of epithelial cells that express a functional TLR3 signaling pathway, we found that TLR3-dependent responses to extracellular dsRNA are negatively regulated by MAVS, precisely "miniMAVS", a recently described 50kDa isoform of MAVS. This regulation of TLR3 by a MAVS isoform constitutes an endogenous regulatory mechanism in epithelial cells that could help prevent a potentially damaging excessive inflammatory response.

    View details for DOI 10.1016/j.cellimm.2016.08.010

    View details for Web of Science ID 000388823700024

    View details for PubMedID 27593154

    View details for PubMedCentralID PMC5125873

  • Epithelial-Derived Inflammation Disrupts Elastin Assembly and Alters Saccutar Stage Lung Development AMERICAN JOURNAL OF PATHOLOGY Benjamin, J. T., van der Meer, R., Im, A. M., Plosa, E. J., Zaynagetdinov, R., Burman, A., Havrilla, M. E., Gleaves, L. A., Polosukhin, V. V., Deutsch, G. H., Yanagisawa, H., Davidson, J. M., Prince, L. S., Young, L. R., Blackwell, T. S. 2016; 186 (7): 1786–1800

    Abstract

    The highly orchestrated interactions between the epithelium and mesenchyme required for normal lung development can be disrupted by perinatal inflammation in preterm infants, although the mechanisms are incompletely understood. We used transgenic (inhibitory κB kinase β transactivated) mice that conditionally express an activator of the NF-κB pathway in airway epithelium to investigate the impact of epithelial-derived inflammation during lung development. Epithelial NF-κB activation selectively impaired saccular stage lung development, with a phenotype comprising rapidly progressive distal airspace dilation, impaired gas exchange, and perinatal lethality. Epithelial-derived inflammation resulted in disrupted elastic fiber organization and down-regulation of elastin assembly components, including fibulins 4 and 5, lysyl oxidase like-1, and fibrillin-1. Fibulin-5 expression by saccular stage lung fibroblasts was consistently inhibited by treatment with bronchoalveolar lavage fluid from inhibitory κB kinase β transactivated mice, Escherichia coli lipopolysaccharide, or tracheal aspirates from preterm infants exposed to chorioamnionitis. Expression of a dominant NF-κB inhibitor in fibroblasts restored fibulin-5 expression after lipopolysaccharide treatment, whereas reconstitution of fibulin-5 rescued extracellular elastin assembly by saccular stage lung fibroblasts. Elastin organization was disrupted in saccular stage lungs of preterm infants exposed to systemic inflammation. Our study reveals a critical window for elastin assembly during the saccular stage that is disrupted by inflammatory signaling and could be amenable to interventions that restore elastic fiber assembly in the developing lung.

    View details for DOI 10.1016/j.ajpath.2016.02.016

    View details for Web of Science ID 000378758400008

    View details for PubMedID 27181406

    View details for PubMedCentralID PMC4929391

  • IL-1 beta and Inflammasome Activity Link Inflammation to Abnormal Fetal Airway Development JOURNAL OF IMMUNOLOGY Stouch, A. N., McCoy, A. M., Greer, R. M., Lakhdari, O., Yull, F. E., Blackwell, T. S., Hoffman, H. M., Prince, L. S. 2016; 196 (8): 3411–20

    Abstract

    Inflammation in the developing preterm lung leads to disrupted airway morphogenesis and chronic lung disease in human neonates. However, the molecular mechanisms linking inflammation and the pathways controlling airway morphogenesis remain unclear. In this article, we show that IL-1β released by activated fetal lung macrophages is the key inflammatory mediator that disrupts airway morphogenesis. In mouse lung explants, blocking IL-1β expression, posttranslational processing, and signaling protected the formation of new airways from the inhibitory effects ofEscherichia coliLPS. Consistent with a critical role for IL-1β, mice expressing a gain-of-functionNlrp3allele and subsequent overactive inflammasome activity displayed abnormal saccular-stage lung morphogenesis and died soon after birth. Although the early-stage fetal lung appeared capable of mounting an NF-κB-mediated immune response, airway formation became more sensitive to inflammation later in development. This period of susceptibility coincided with higher expression of multiple inflammasome components that could increase the ability to release bioactive IL-1β. Macrophages fromNlrp3gain-of-function mice also expressed higher levels of more mature cell surface markers, additionally linking inflammasome activation with macrophage maturation. These data identify developmental expression of the inflammasome and IL-1β release by fetal lung macrophages as key mechanisms and potential therapeutic targets for neonatal lung disease.

    View details for DOI 10.4049/jimmunol.1500906

    View details for Web of Science ID 000373721900019

    View details for PubMedID 26951798

    View details for PubMedCentralID PMC5315059

  • Molecular Imaging of Folate Receptor beta-Positive Macrophages during Acute Lung Inflammation AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY Han, W., Zaynagetdinov, R., Yull, F. E., Polosukhin, V. V., Gleaves, L. A., Tanjore, H., Young, L. R., Peterson, T. E., Manning, H., Prince, L. S., Blackwell, T. S. 2015; 53 (1): 50–59

    Abstract

    Characterization of markers that identify activated macrophages could advance understanding of inflammatory lung diseases and facilitate development of novel methodologies for monitoring disease activity. We investigated whether folate receptor β (FRβ) expression could be used to identify and quantify activated macrophages in the lungs during acute inflammation induced by Escherichia coli LPS. We found that FRβ expression was markedly increased in lung macrophages at 48 hours after intratracheal LPS. In vivo molecular imaging with a fluorescent probe (cyanine 5 polyethylene glycol folate) showed that the fluorescence signal over the chest peaked at 48 hours after intratracheal LPS and was markedly attenuated after depletion of macrophages. Using flow cytometry, we identified the cells responsible for uptake of cyanine 5-conjugated folate as FRβ(+) interstitial macrophages and pulmonary monocytes, which coexpressed markers associated with an M1 proinflammatory macrophage phenotype. These findings were confirmed using a second model of acute lung inflammation generated by inducible transgenic expression of an NF-κB activator in airway epithelium. Using CC chemokine receptor 2-deficient mice, we found that FRβ(+) macrophage/monocyte recruitment was dependent on the monocyte chemotactic protein-1/CC chemokine receptor 2 pathway. Together, our results demonstrate that folate-based molecular imaging can be used as a noninvasive approach to detect classically activated monocytes/macrophages recruited to the lungs during acute inflammation.

    View details for DOI 10.1165/rcmb.2014-0289OC

    View details for Web of Science ID 000362125500008

    View details for PubMedID 25375039

    View details for PubMedCentralID PMC4566110

  • Epithelial beta 1 integrin is required for lung branching morphogenesis and alveolarization DEVELOPMENT Plosa, E. J., Young, L. R., Gulleman, P. M., Polosukhin, V. V., Zaynagetdinov, R., Benjamin, J. T., Im, A. M., van der Meer, R., Gleaves, L. A., Bulus, N., Han, W., Prince, L. S., Blackwell, T. S., Zent, R. 2014; 141 (24): 4751–62

    Abstract

    Integrin-dependent interactions between cells and extracellular matrix regulate lung development; however, specific roles for β1-containing integrins in individual cell types, including epithelial cells, remain incompletely understood. In this study, the functional importance of β1 integrin in lung epithelium during mouse lung development was investigated by deleting the integrin from E10.5 onwards using surfactant protein C promoter-driven Cre. These mutant mice appeared normal at birth but failed to gain weight appropriately and died by 4 months of age with severe hypoxemia. Defects in airway branching morphogenesis in association with impaired epithelial cell adhesion and migration, as well as alveolarization defects and persistent macrophage-mediated inflammation were identified. Using an inducible system to delete β1 integrin after completion of airway branching, we showed that alveolarization defects, characterized by disrupted secondary septation, abnormal alveolar epithelial cell differentiation, excessive collagen I and elastin deposition, and hypercellularity of the mesenchyme occurred independently of airway branching defects. By depleting macrophages using liposomal clodronate, we found that alveolarization defects were secondary to persistent alveolar inflammation. β1 integrin-deficient alveolar epithelial cells produced excessive monocyte chemoattractant protein 1 and reactive oxygen species, suggesting a direct role for β1 integrin in regulating alveolar homeostasis. Taken together, these studies define distinct functions of epithelial β1 integrin during both early and late lung development that affect airway branching morphogenesis, epithelial cell differentiation, alveolar septation and regulation of alveolar homeostasis.

    View details for DOI 10.1242/dev.117200

    View details for Web of Science ID 000346431900011

    View details for PubMedID 25395457

    View details for PubMedCentralID PMC4299273

  • Epithelial-mesenchymal co-culture model for studying alveolar morphogenesis ORGANOGENESIS Greer, R. M., Miller, J., Okoh, V. O., Halloran, B. A., Prince, L. S. 2014; 10 (4): 340–49

    Abstract

    Division of large, immature alveolar structures into smaller, more numerous alveoli increases the surface area available for gas exchange. Alveolar division requires precise epithelial-mesenchymal interactions. However, few experimental models exist for studying how these cell-cell interactions produce changes in 3-dimensional structure. Here we report an epithelial-mesenchymal cell co-culture model where 3-dimensional peaks form with similar cellular orientation as alveolar structures in vivo. Co-culturing fetal mouse lung mesenchyme with A549 epithelial cells produced tall peaks of cells covered by epithelia with cores of mesenchymal cells. These structures did not form when using adult lung fibroblasts. Peak formation did not require localized areas of cell proliferation or apoptosis. Mesenchymal cells co-cultured with epithelia adopted an elongated cell morphology closely resembling myofibroblasts within alveolar septa in vivo. Because inflammation inhibits alveolar formation, we tested the effects of E. coli lipopolysaccharide on 3-dimensional peak formation. Confocal and time-lapse imaging demonstrated that lipopolysaccharide reduced mesenchymal cell migration, resulting in fewer, shorter peaks with mesenchymal cells present predominantly at the base. This epithelial-mesenchymal co-culture model may therefore prove useful in future studies of mechanisms regulating alveolar morphogenesis.

    View details for DOI 10.4161/org.29198

    View details for Web of Science ID 000352611600002

    View details for PubMedID 25482312

    View details for PubMedCentralID PMC4594603

  • I kappa B Kinase Activity Drives Fetal Lung Macrophage Maturation along a Non-M1/M2 Paradigm JOURNAL OF IMMUNOLOGY Stouch, A. N., Zaynagetdinov, R., Barham, W. J., Stinnett, A. M., Slaughter, J. C., Yull, F. E., Hoffman, H. M., Blackwell, T. S., Prince, L. S. 2014; 193 (3): 1184–93

    Abstract

    In preterm infants, exposure to inflammation increases the risk of bronchopulmonary dysplasia, a chronic, developmental lung disease. Although macrophages are the key cells that initiate lung inflammation, less is known about lung macrophage phenotype and maturation. We hypothesized that fetal lung macrophages mature into distinct subpopulations during mouse development, and that activation could influence macrophage maturation. Expression of the fetal macrophage markers CD68, CD86, CD206, Ym1, fibrinogen-like protein 2, and indolamine-2, 3-dioxygenase was developmentally regulated, with each marker having different temporal patterns. Flow cytometry analysis showed macrophages within the fetal lung were less diverse than the distinctly separate subpopulations in newborn and adult lungs. Similar to adult alveolar macrophages, fetal lung macrophages responded to the TLR4 agonist LPS and the alternative activation cytokines IL-4 and IL-13. Using a macrophage-specific constitutively active IκB Kinase transgenic model (IKFM), we demonstrated that macrophage activation increased proinflammatory gene expression and reduced the response of fetal lung macrophages to IL-4 and IL-13. Activation also increased fetal lung macrophage proliferation. Fetal IKFM lungs contained increased percentages of more mature, CD11b(low)F4/80(high) cells that also expressed higher levels of the alternative activation markers CD204 and CD206. Development of fetal lung macrophages into mature alveolar macrophages may therefore include features of both proinflammatory and alternative activation paradigms.

    View details for DOI 10.4049/jimmunol.1302516

    View details for Web of Science ID 000339422000024

    View details for PubMedID 24981452

    View details for PubMedCentralID PMC4108541

  • Epithelial-mesenchymal co-culture model for studying alveolar morphogenesis. Organogenesis Greer, R. M., Miller, J. D., Okoh, V. O., Halloran, B. A., Prince, L. S. 2014; 10 (3)

    Abstract

    Division of large, immature alveolar structures into smaller, more numerous alveoli increases the surface area available for gas exchange. Alveolar division requires precise epithelial-mesenchymal interactions. However, few experimental models exist for studying how these cell-cell interactions produce changes in 3-dimensional structure. Here we report an epithelial-mesenchymal cell co-culture model where 3-dimensional peaks form with similar cellular orientation as alveolar structures in vivo. Co-culturing fetal mouse lung mesenchyme with A549 epithelial cells produced tall peaks of cells covered by epithelia with cores of mesenchymal cells. These structures did not form when using adult lung fibroblasts. Peak formation did not require localized areas of cell proliferation or apoptosis. Mesenchymal cells co-cultured with epithelia adopted an elongated cell morphology closely resembling myofibroblasts within alveolar septa in vivo. Because inflammation inhibits alveolar formation, we tested the effects of E. coli lipopolysaccharide on 3-dimensional peak formation. Confocal and time-lapse imaging demonstrated that lipopolysaccharide reduced mesenchymal cell migration, resulting in fewer, shorter peaks with mesenchymal cells present predominantly at the base. This epithelial-mesenchymal co-culture model may therefore prove useful in future studies of mechanisms regulating alveolar morphogenesis.

    View details for PubMedID 24827973

  • Molecular determinants of lung development. Annals of the American Thoracic Society Morrisey, E. E., Cardoso, W. V., Lane, R. H., Rabinovitch, M., Abman, S. H., Ai, X., Albertine, K. H., Bland, R. D., Chapman, H. A., Checkley, W., Epstein, J. A., Kintner, C. R., Kumar, M., Minoo, P., Mariani, T. J., McDonald, D. M., Mukouyama, Y., Prince, L. S., Reese, J., Rossant, J., Shi, W., Sun, X., Werb, Z., Whitsett, J. A., Gail, D., Blaisdell, C. J., Lin, Q. S. 2013; 10 (2): S12-6

    Abstract

    Development of the pulmonary system is essential for terrestrial life. The molecular pathways that regulate this complex process are beginning to be defined, and such knowledge is critical to our understanding of congenital and acquired lung diseases. A recent workshop was convened by the National Heart, Lung, and Blood Institute to discuss the developmental principles that regulate the formation of the pulmonary system. Emerging evidence suggests that key developmental pathways not only regulate proper formation of the pulmonary system but are also reactivated upon postnatal injury and repair and in the pathogenesis of human lung diseases. Molecular understanding of early lung development has also led to new advances in areas such as generation of lung epithelium from pluripotent stem cells. The workshop was organized into four different topics, including early lung cell fate and morphogenesis, mechanisms of lung cell differentiation, tissue interactions in lung development, and environmental impact on early lung development. Critical points were raised, including the importance of epigenetic regulation of lung gene expression, the dearth of knowledge on important mesenchymal lineages within the lung, and the interaction between the developing pulmonary and cardiovascular system. This manuscript describes the summary of the discussion along with general recommendations to overcome the gaps in knowledge in lung developmental biology.

    View details for DOI 10.1513/AnnalsATS.201207-036OT

    View details for PubMedID 23607856