Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Curr Allergy Asthma Rep ; 23(2): 77-91, 2023 02.
Article in English | MEDLINE | ID: mdl-36609951

ABSTRACT

PURPOSE OF REVIEW: There has been an increased prevalence of allergy. Due to this relatively rapid rise, changes in environmental exposures are likely the main contributor. In this review, we highlight literature from the last 3 years pertaining to the role of air pollution, greenness, and the rural/farm lifestyle and their association with the development of allergic sensitization, atopic dermatitis, food allergy, and allergic rhinitis in infancy and childhood. Because asthma has a more complex pathophysiology, it was excluded from this review. RECENT FINDINGS: Recent studies support a role for air pollution, greenness, and rural/farming lifestyle influencing atopic outcomes that continue to be defined. While many studies have examined singular environmental exposures, the interconnectedness of these exposures and others points to a need for future work to consider an individual's whole exposure. Environmental exposures' influence on atopic disease development remains an ongoing and important area of study.


Subject(s)
Air Pollution , Dermatitis, Atopic , Rhinitis, Allergic , Humans , Child , Farms , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/etiology , Air Pollution/adverse effects , Rhinitis, Allergic/epidemiology , Environmental Exposure/adverse effects
3.
Front Pediatr ; 10: 916184, 2022.
Article in English | MEDLINE | ID: mdl-35874571

ABSTRACT

Traditional farming lifestyle has been shown to be protective against asthma and allergic diseases. The individual factors that appear to be associated with this "farm-life effect" include consumption of unpasteurized farm milk and exposure to farm animals and stables. However, the biomarkers of the protective immunity and those associated with early development of allergic diseases in infancy remain unclear. The "Zooming in to Old Order Mennonites (ZOOM)" study was designed to assess the differences in the lifestyle and the development of the microbiome, systemic and mucosal immunity between infants born to traditional farming lifestyle at low risk for allergic diseases and those born to urban/suburban atopic families with a high risk for allergic diseases in order to identify biomarkers of development of allergic diseases in infancy. 190 mothers and their infants born to Old Order Mennonite population protected from or in Rochester families at high risk for allergic diseases were recruited before birth from the Finger Lakes Region of New York State. Questionnaires and samples are collected from mothers during pregnancy and after delivery and from infants at birth and at 1-2 weeks, 6 weeks, 6, 12, 18, and 24 months, with 3-, 4-, and 5-year follow-up ongoing. Samples collected include maternal blood, stool, saliva, nasal and skin swabs and urine during pregnancy; breast milk postnatally; infant blood, stool, saliva, nasal and skin swabs. Signs and symptoms of allergic diseases are assessed at every visit and serum specific IgE is measured at 1 and 2 years of age. Allergic diseases are diagnosed by clinical history, exam, and sensitization by skin prick test and/or serum specific IgE. By the end of the first year of life, the prevalence of food allergy and atopic dermatitis were higher in ROC infants compared to the rates observed in OOM infants as was the number of infants sensitized to foods. These studies of immune system development in a population protected from and in those at risk for allergic diseases will provide critical new knowledge about the development of the mucosal and systemic immunity and lay the groundwork for future studies of prevention of allergic diseases.

4.
Ann Allergy Asthma Immunol ; 129(3): 292-300, 2022 09.
Article in English | MEDLINE | ID: mdl-35490857

ABSTRACT

OBJECTIVE: To review existing literature on the early risk factors for and biomarkers of food allergy (FA) and food sensitization (FS) and highlight opportunities for future research that will further the understanding of FA pathogenesis in infancy and toddlerhood. DATA SOURCES: PubMed search of English-language articles related to FA and atopic disease. STUDY SELECTIONS: Human studies with outcomes related to FA, FS, and other atopic disease in childhood were selected and reviewed. Studies published after 2015 were prioritized. RESULTS: The prevalence of FA has greatly increased in recent decades and is now a global public health concern. A complex network of early life risk factors has been associated with development of FA and FS in childhood. Food allergy has a genetic component, but recent evidence suggests that interactions between risk alleles and other environmental exposures are important for disease pathogenesis, potentially through epigenetic mechanisms. Lifestyle factors, such as delivery mode, antibiotic use, and pet exposure also influence FA risk, which may be through their effect on the early life gut microbiome. How these early life risk factors, along with route and timing of antigen exposure, collectively target the developing immune system remains an ongoing and important area of study. CONCLUSION: The current body of evidence emphasizes the first 1000 days of life as a critical period for FA development. More observational studies and adequately powered clinical trials spanning early pregnancy through childhood are needed to identify novel biomarkers and risk factors that can predict susceptibility toward or protection against FA.


Subject(s)
Food Hypersensitivity , Allergens , Biomarkers , Child, Preschool , Environmental Exposure/adverse effects , Epigenesis, Genetic , Female , Food Hypersensitivity/etiology , Humans , Pregnancy
5.
Sci Transl Med ; 14(638): eabl8574, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35353543

ABSTRACT

Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1ß and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.


Subject(s)
Chorioamnionitis , Premature Birth , Animals , Chorioamnionitis/chemically induced , Chorioamnionitis/pathology , Female , Lung/pathology , Macaca mulatta , Pregnancy , Premature Birth/prevention & control , Pulmonary Gas Exchange
6.
Mucosal Immunol ; 15(4): 730-744, 2022 04.
Article in English | MEDLINE | ID: mdl-35314757

ABSTRACT

Up to 40% of preterm births are associated with histological chorioamnionitis (HCA), which leads to elevated levels of pro-inflammatory mediators and microbial products in the amniotic fluid, which come in contact with fetal lungs. Yet, fetal pulmonary immune responses to such exposure remain poorly characterized. To address this gap, we used our established HCA model, in which pregnant Rhesus macaques receive intraamniotic (IA) saline or LPS. IA LPS induced a potent and rapid myeloid cell response in fetal lungs, dominated by neutrophils and monocytes/macrophages. Infiltrating and resident myeloid cells exhibited transcriptional profiles consistent with exposure to TLR ligands, as well as cytokines, notably IL-1 and TNFα. Although simultaneous, in vivo blockade of IL-1 and TNFα signaling did not prevent the inflammatory cell recruitment, it blunted the lung overall inflammatory state reducing communication between, and activation of, infiltrating immune cells. Our data indicate that the fetal innate immune system can mount a rapid multi-faceted pulmonary immune response to in utero exposure to inflammation. These data provide mechanistic insights into the association between HCA and the postnatal lung morbidities of the premature infant and highlight therapeutic potential of inflammatory blockade in the fetus.


Subject(s)
Chorioamnionitis , Pneumonia , Premature Birth , Amniotic Fluid , Animals , Chorioamnionitis/pathology , Female , Humans , Inflammation , Interleukin-1 , Lipopolysaccharides , Lung , Macaca mulatta , Pregnancy , Premature Birth/pathology , Tumor Necrosis Factor-alpha
7.
Acta Paediatr ; 111(1): 54-58, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34626494

ABSTRACT

There has been an increased prevalence of several allergic manifestations such as food allergy, atopic eczema, allergic rhinitis and asthma. Several explanations have been proposed why this has occurred, but one of the main contributing factors may be the gradual loss of microbial exposures over time in regions where allergy is prevalent. Such exposures occur in individuals who practise a traditional farming lifestyle and are protected against allergy. Infant consumption of human milk, more commonly practised in these farming communities, may provide an alternative in combatting allergy, as it known to be beneficial to infant health. In this review, we cover human milk and its role in shaping the gut microbiome promoting the growth of beneficial bacteria like Bifidobacterium, as well as the downstream impact of the farming lifestyle, human milk and Bifidobacterium has on developing infant immunity.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Agriculture , Humans , Infant , Life Style , Milk, Human
8.
JCI Insight ; 5(24)2020 12 17.
Article in English | MEDLINE | ID: mdl-33328385

ABSTRACT

Respiratory complicˆations are the major cause of morbidity and mortality among preterm infants, which is partially prevented by the administration of antenatal corticosteroids (ACS). Most very preterm infants are exposed to chorioamnionitis, but short- and long-term effects of ACS treatment in this setting are not well defined. In low-resource settings, ACS increased neonatal mortality by perhaps increasing infection. We report that treatment with low-dose ACS in the setting of inflammation induced by intraamniotic lipopolysaccharide (LPS) in rhesus macaques improves lung compliance and increases surfactant production relative to either exposure alone. RNA sequencing shows that these changes are mediated by suppression of proliferation and induction of mesenchymal cellular death via TP53. The combined exposure results in a mature-like transcriptomic profile with inhibition of extracellular matrix development by suppression of collagen genes COL1A1, COL1A2, and COL3A1 and regulators of lung development FGF9 and FGF10. ACS and inflammation also suppressed signature genes associated with proliferative mesenchymal progenitors similar to the term gestation lung. Treatment with ACS in the setting of inflammation may result in early respiratory advantage to preterm infants, but this advantage may come at a risk of abnormal extracellular matrix development, which may be associated with increased risk of chronic lung disease.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Lung/drug effects , Premature Birth/drug therapy , Adrenal Cortex Hormones/adverse effects , Animals , Animals, Newborn , Chorioamnionitis/drug therapy , Chorioamnionitis/genetics , Dexamethasone/pharmacology , Disease Models, Animal , Female , Glucocorticoids/pharmacology , Inflammation/drug therapy , Inflammation/genetics , Macaca mulatta , Male , Pregnancy , Pulmonary Surfactants/pharmacology
9.
Front Immunol ; 11: 1285, 2020.
Article in English | MEDLINE | ID: mdl-32636848

ABSTRACT

Chorioamnionitis, a potentially serious inflammatory complication of pregnancy, is associated with the development of an inflammatory milieu within the amniotic fluid surrounding the developing fetus. When chorioamnionitis occurs, the fetal lung finds itself in the unique position of being constantly exposed to the consequent inflammatory meditators and/or microbial products found in the amniotic fluid. This exposure results in significant changes to the fetal lung, such as increased leukocyte infiltration, altered cytokine, and surfactant production, and diminished alveolarization. These alterations can have potentially lasting impacts on lung development and function. However, studies to date have only begun to elucidate the association between such inflammatory exposures and lifelong consequences such as lung dysfunction. In this review, we discuss the pathogenesis of and fetal immune response to chorioamnionitis, detail the consequences of chorioamnionitis exposure on the developing fetal lung, highlighting the various animal models that have contributed to our current understanding and discuss the importance of fetal exposures in regard to the development of chronic respiratory disease. Finally, we focus on the clinical, basic, and therapeutic challenges in fetal inflammatory injury to the lung, and propose next steps and future directions to improve our therapeutic understanding of this important perinatal stress.


Subject(s)
Chorioamnionitis/immunology , Fetus/immunology , Lung/embryology , Lung/pathology , Prenatal Exposure Delayed Effects/immunology , Animals , Chorioamnionitis/pathology , Female , Humans , Pregnancy , Prenatal Exposure Delayed Effects/pathology
10.
Front Immunol ; 11: 558, 2020.
Article in English | MEDLINE | ID: mdl-32308656

ABSTRACT

Accumulation of activated neutrophils at the feto-maternal interface is a defining hallmark of intrauterine inflammation (IUI) that might trigger an excessive immune response during pregnancy. Mechanisms responsible of this massive neutrophil recruitment are poorly investigated. We have previously showed that intraamniotic injection of LPS in rhesus macaques induced a neutrophil predominant inflammatory response similar to that seen in human IUI. Here, we demonstrate that anti-TNF antibody (Adalimumab) inhibited ~80% of genes induced by LPS involved in inflammatory signaling and innate immunity in chorio-decidua neutrophils. Consistent with the gene expression data, TNF-blockade decreased LPS-induced neutrophil accumulation and activation at the feto-maternal interface. We also observed a reduction in IL-6 and other pro-inflammatory cytokines but not prostaglandins concentrations in the amniotic fluid. Moreover, TNF-blockade decreased mRNA expression of inflammatory cytokines in the chorio-decidua but not in the uterus, suggesting that inhibition of TNF-signaling decreased the inflammation in a tissue-specific manner within the uterine compartment. Taken together, our results demonstrate a predominant role for TNF-signaling in modulating the neutrophilic infiltration at the feto-maternal interface during IUI and suggest that blockade of TNF-signaling could be considered as a therapeutic approach for IUI, the major leading cause of preterm birth.


Subject(s)
Chorioamnionitis/immunology , Neutrophils/immunology , Tumor Necrosis Factor-alpha/immunology , Adalimumab/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Chorioamnionitis/chemically induced , Female , Lipopolysaccharides/toxicity , Macaca mulatta , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Pregnancy , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
J Lipid Res ; 58(8): 1514-1523, 2017 08.
Article in English | MEDLINE | ID: mdl-28377425

ABSTRACT

HDLs appear to affect regulatory T cell (Treg) homeostasis, as suggested by the increased Treg counts in HDL-treated mice and by the positive correlation between Treg frequency and HDL-cholesterol levels in statin-treated healthy adults. However, the underlying mechanisms remain unclear. Herein, we show that HDLs, not LDLs, significantly decreased the apoptosis of human Tregs in vitro, whereas they did not alter naïve or memory CD4+ T cell survival. Similarly, oleic acid bound to serum albumin increased Treg survival. Tregs bound and internalized high amounts of HDL compared with other subsets, which might arise from the higher expression of the scavenger receptor class B type I by Tregs; accordingly, blocking this receptor hindered HDL-mediated Treg survival. Mechanistically, we showed that HDL increased Treg ATP concentration and mitochondrial activity, enhancing basal respiration, maximal respiration, and spare respiratory capacity. Blockade of FA oxidation by etoxomir abolished the HDL-mediated enhanced survival and mitochondrial activity. Our findings thus suggest that Tregs can specifically internalize HDLs from their microenvironment and use them as an energy source. Furthermore, a novel implication of our data is that enhanced Treg survival may contribute to HDLs' anti-inflammatory properties.


Subject(s)
Lipoproteins, HDL/metabolism , T-Lymphocytes, Regulatory/cytology , Adenosine Triphosphate/biosynthesis , CD36 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Fatty Acids/metabolism , Homeostasis , Humans , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , T-Lymphocytes, Regulatory/metabolism
12.
Sci Rep ; 7(1): 465, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28352109

ABSTRACT

Differentiation of regulatory Treg (Treg) in the periphery is critical to control inflammatory processes. Although polarization of inducible Treg (iTreg) often occurs in an inflammatory environment, the effects exerted by inflammation on human iTreg differentiation have not been extensively studied. We observed that IL-1ß significantly reduced the frequency of FOXP3+ T cells under iTreg-polarizing conditions. Mechanistically, we show that IL-1ß activated mTORC1 and downstream upregulated hypoxia inducible factor-1 (HIF-1α) expression. Using specific inhibitors, we demonstrated that both steps were critical in the deleterious effect of IL-1ß on Treg differentiation. Chemical stabilization of HIF-1α by Dimethyloxalylglycine (DMOG) also significantly impaired iTreg differentiation. Interestingly, while IL-1ß-treated cells exhibited only minor changes in metabolism, DMOG treatment decreased iTreg mitochondrial respiration and increased their glycolytic capacity. In conclusion, exposure to inflammatory stimuli profoundly inhibits human Treg differentiation HIF-1α dependent, suggesting that targeting HIF-1α could be a strategy to foster iTreg differentiation in an inflammatory milieu. However, IL-1ß deleterious effect does not appear to be completely driven by metabolic changes. These data thus suggest that several mechanisms contribute to the regulation of iTreg differentiation, but the timing and respective requirement for each pathway vary depending on the milieu in which iTreg differentiate.


Subject(s)
Cell Differentiation , Forkhead Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Amino Acids, Dicarboxylic/metabolism , Biomarkers , Cell Differentiation/drug effects , Cell Respiration , Gene Expression , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunophenotyping , Mitochondria/genetics , Mitochondria/metabolism , Phenotype , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism
13.
Pediatr Res ; 81(2): 384-390, 2017 02.
Article in English | MEDLINE | ID: mdl-27814345

ABSTRACT

BACKGROUND: Acute chorioamnionitis contributes to premature birth, and is associated with postbirth complications. How chorioamnionitis impacts neonate's developing immune system has not been well defined. METHODS: Blood from extremely preterm infants (≤28 wk gestation) was drawn at the first, second, and fourth week of life. Blood was either left unstimulated or stimulated for 4 h with PMA/ionomycin. mRNA expression of transcription factors in unstimulated cells (RORC, TBET, GATA3, and Forkhead box protein 3 (FOXP3)) and inflammatory cytokines (IFN-γ, TNF-α, IL-2, IL-4, IL-5, and IL-6) in unstimulated and stimulated cells were analyzed. Data were analyzed based on the diagnosis of chorioamnionitis, funisitis and bronchopulmonary dysplasia (BPD). RESULTS: At 1 wk of life, exposure to funisitis, but not maternal chorioamnionitis was associated with an increased expression of RORC and RORC/FOXP3 ratio. These increases in RORC and RORC/FOXP3 ratio were sustained over the 4 wk of follow-up. Leukocytes from infants who developed BPD had increased stimulated and unstimulated IL-4 at the first week of life, but these increases were not sustained over time. In contrast, infants with mild BPD had a sustained decrease in stimulated IL-2. CONCLUSION: Chorioamnionitis exposure, in particular to funisitis, lead to enhanced Th17-like responses that persist for 4 wk after birth. Infants who later developed BPD did not exhibit a strikingly distinct immune profile.


Subject(s)
Bronchopulmonary Dysplasia/immunology , Chorioamnionitis/immunology , Bronchopulmonary Dysplasia/blood , Chorioamnionitis/blood , Cytokines/blood , Female , Gene Expression Regulation , Humans , Infant, Newborn , Infant, Premature , Inflammation , Male , Pregnancy , Th17 Cells/cytology , Transcription Factors/blood
14.
Front Immunol ; 7: 216, 2016.
Article in English | MEDLINE | ID: mdl-27313580

ABSTRACT

Regulatory T-cells (Tregs) mediate their suppressive action by acting directly on conventional T-cells (Tcons) or dendritic cells (DCs). One mechanism of Treg suppression is the increase of cyclic adenosine 3',5'-monophosphate (cAMP) levels in target cells. Tregs utilize cAMP to control Tcon responses, such as proliferation and cytokine production. Tregs also exert their suppression on DCs, diminishing DC immunogenicity by downmodulating the expression of costimulatory molecules and actin polymerization at the immunological synapse. The Treg-mediated usage of cAMP occurs through two major mechanisms. The first involves the Treg-mediated influx of cAMP in target cells through gap junctions. The second is the conversion of adenosine triphosphate into adenosine by the ectonucleases CD39 and CD73 present on the surface of Tregs. Adenosine then binds to receptors on the surface of target cells, leading to increased intracellular cAMP levels in these targets. Downstream, cAMP can activate the canonical protein kinase A (PKA) pathway and the exchange protein activated by cyclic AMP (EPAC) non-canonical pathway. In this review, we discuss the most recent findings related to cAMP activation of PKA and EPAC, which are implicated in Treg homeostasis as well as the functional alterations induced by cAMP in cellular targets of Treg suppression.

15.
J Immunol ; 196(9): 3706-15, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27036917

ABSTRACT

Chorioamnionitis is associated with preterm labor and fetal inflammatory response syndrome (FIRS), causing fetal organ injury and morbidity, particularly in extremely premature infants. However, the effects of inflammation on the fetal immune system remain poorly understood, due to the difficulty of studying immune development in infants. Therefore, we used the model of intra-amniotic LPS administered at ∼80% gestation in rhesus monkeys to cause chorioamnionitis and FIRS that is similar in human pathology. Importantly, the frequency of IL-17(+) and IL-22(+) CD4(+) T cells increased in the spleen of LPS-exposed fetuses, whereas regulatory T cell (Treg) frequency decreased. These changes persisted for at least 48 h. Notably, Th17 cytokines were predominantly expressed by FOXP3(+)CD4(+) T cells and not by their FOXP3(-) counterparts. Bifunctional IL-17(+)FOXP3(+) exhibited a phenotype of inflammatory Tregs (RORc(High/+), Helios(Low/-), IL-2(+), IFN-γ(+), and IL-8(+)) compared with typical FOXP3(+) cells. Diminished splenic Treg frequency in LPS-exposed fetuses was associated with inadequate Treg generation in the thymus. Mechanistically, the emergence of inflammatory Tregs was largely dependent on IL-1 signaling. However, blockage of IL-1R signaling did not abolish the deleterious effects of LPS on Treg frequency in the thymus or spleen. Collectively, we demonstrate that a prenatal inflammatory environment leads to inadequate Treg generation in the thymus with a switch of splenic Tregs toward an inflammatory phenotype. Both processes likely contribute to the pathogenesis of chorioamnionitis. Approaches to manipulate Treg numbers and function could thus be useful therapeutically to alleviate FIRS in preterm infants.


Subject(s)
Chorioamnionitis/immunology , Immunotherapy/trends , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Interleukin-1/metabolism , Obstetric Labor, Premature/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Chorioamnionitis/therapy , Disease Models, Animal , Female , Fetus , Forkhead Transcription Factors/metabolism , Humans , Lipopolysaccharides/immunology , Macaca mulatta , Obstetric Labor, Premature/therapy , Pregnancy , Signal Transduction
16.
Eur J Immunol ; 45(9): 2582-92, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26046326

ABSTRACT

Regulatory T cells (Treg cells) limit contact between dendritic cells (DCs) and conventional T cells (Tcons), decreasing the formation of aggregates as well as down-modulating the expression of co-stimulatory molecules by DCs, thus decreasing DC immunogenicity and abrogating T-cell activation. Despite the importance of this Treg-cell function, the capacity of Treg cells from term and preterm neonates to suppress DCs, and the suppressive mechanisms they use, are still undefined. We found that, relative to adult Treg cells, activated Treg cells from human neonates expressed lower FOXP3 and CTLA-4, but contained higher levels of cAMP. We developed an in vitro model in which Treg function was measured at a physiological ratio of 1 Treg for 10 Tcon and 1 monocyte-derived DC, as Treg target. Term and preterm Treg cells failed to suppress the formation of DC-Tcon aggregates, in contrast to naïve and memory Treg cells from adults. However, neonatal Treg cells diminished DC and Tcon activation as well as actin polymerization at the immunological synapses. In addition, CTLA-4 and cAMP were the main suppressive molecules used by neonatal Treg. Altogether, both preterm and term neonatal Treg cells appear less functional than adult Treg cells, and this defect is consistent with the general impairment of CD4 cell function in newborns.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/immunology , Actins/chemistry , Actins/genetics , Actins/immunology , Adult , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Aggregation/immunology , Cell Separation , Cyclic AMP/immunology , Cyclic AMP/metabolism , Dendritic Cells/cytology , Female , Fetal Blood/cytology , Fetal Blood/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Humans , Immunological Synapses/chemistry , Immunological Synapses/metabolism , Infant, Newborn , Infant, Premature , Male , Signal Transduction , T-Lymphocytes, Regulatory/cytology
17.
Stem Cells Dev ; 24(10): 1236-51, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25567291

ABSTRACT

Multi-drug resistance (MDR)-ATP binding cassette (ABC) transporters, ABCB1, ABCC1, and ABCG2 participate in the efflux of steroid hormones, estrogens, and androgens, which regulate prostate development and differentiation. The role of MDR-ABC efflux transporters in prostate epithelial proliferation and differentiation remains unclear. We hypothesized that MDR-ABC transporters regulate prostate differentiation and epithelium regeneration. Prostate epithelial differentiation was studied using histology, sphere formation assay, and prostate regeneration induced by cycles of repeated androgen withdrawal and replacement. Embryonic deletion of Abcg2 resulted in a decreased number of luminal cells in the prostate and increased sphere formation efficiency, indicating an imbalance in the prostate epithelial differentiation pattern. Decreased luminal cell number in the Abcg2 null prostate implies reduced differentiation. Enhanced sphere formation efficiency in Abcg2 null prostate cells implies activation of the stem/progenitor cells. Prostate regeneration was associated with profound activation of the stem/progenitor cells, indicating the role of Abcg2 in maintaining stem/progenitor cell pool. Since embryonic deletion of Abcg2 may result in compensation by other ABC transporters, pharmacological inhibition of MDR-ABC efflux was performed. Pharmacological inhibition of MDR-ABC efflux enhanced prostate epithelial differentiation in sphere culture and during prostate regeneration. In conclusion, Abcg2 deletion leads to activation of the stem/progenitor cells and enhances differentiating divisions; and pharmacological inhibition of MDR-ABC efflux leads to epithelial differentiation. Our study demonstrates for the first time that MDR-ABC efflux transporter inhibition results in enhanced prostate epithelial cell differentiation.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Cell Differentiation/physiology , Drug Resistance, Multiple , Prostate/cytology , Stem Cells/cytology , Animals , Cell Line, Tumor , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Female , Male , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/metabolism , Prostate/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...