Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Placenta ; 142: 1-11, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37579594

ABSTRACT

INTRODUCTION: Maternal prenatal psychological stress is associated with adverse pregnancy outcomes and increased risk of adverse health outcomes in children. While the molecular mechanisms that govern these associations has not been fully teased apart, stress-induced changes in placental function can drive sex-specific phenotypes in offspring. We sought to identify and examine molecular pathways in the placenta that are altered in response to maternal prenatal stress. METHODS: We previously employed a mouse model of maternal prenatal stress where pregnant dams were treated with stress hormone (CORT) beginning in mid-gestation. Using this model, we conducted RNAseq analysis of whole placenta at E18.5. We used qRT-PCR to validate gene expression changes in the placenta and in a trophoblast cell line. ELISAs were used to measure the abundance of thyroid hormones in maternal and fetal serum and in the placenta. RESULTS: Dio2 was amongst the top differentially expressed genes in response to exogenous stress hormone. Dio2 expression was more downregulated in placenta of female fetuses from CORT-treated dams than both control placenta from females and placenta from male fetuses. Consistent with Dio2's role in production of bioactive thyroid hormone (T3), we found that there was a reduction of T3 in placenta and serum of female embryos from CORT-treated dams at E18.5. Both T3 and T4 were reduced in the fetal compartment of the placenta of female fetuses from CORT-treated dams at E16.5. Exogenous stress hormone induced reduction in thyroid hormone in females was independent of circulating levels of TH in the dams. DISCUSSION: The placental thyroid hormone synthesis pathway may be a target of elevated maternal stress hormone and modulate fetal programming of health and disease of offspring in a sex-specific fashion.


Subject(s)
Corticosterone , Placenta , Humans , Child , Pregnancy , Female , Male , Mice , Animals , Placenta/metabolism , Corticosterone/pharmacology , Corticosterone/metabolism , Thyroid Hormones , Fetus/metabolism , Thyroid Gland
2.
bioRxiv ; 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37461599

ABSTRACT

Introduction: Maternal prenatal stress is associated with adverse pregnancy outcomes and predisposition to long-term adverse health outcomes in children. While the molecular mechanisms that govern these associations has not been fully teased apart, stress-induced changes in placental function can drive sex-specific phenotypes in offspring. We sought to identify and examine molecular pathways in the placenta that are altered in response to maternal prenatal stress. Methods: Using a mouse model of maternal prenatal stress, we conducted RNA-seq analysis of whole placenta at E18.5. We used qRT-PCR to validate gene expression changes in the placenta and in a trophoblast cell line. ELISAs were used to measure the abundance of thyroid hormones in maternal and fetal serum and in the placenta. Results: Dio2 was amongst the top differentially expressed genes in response to elevated maternal stress hormone. Dio2 expression was more downregulated in female placenta from stressed dams than both female control and male placenta. Consistent with Dio2's role in production of bioactive thyroid hormone (T3), we found that there was a reduction of T3 in placenta and serum of female embryos from stressed dams at E18.5. Both T3 and T4 were reduced in the fetal compartment of the female placenta from stressed dams at E16.5. Stress hormone induced reduction in thyroid hormone in females was independent of circulating levels of TH in the dams. Discussion: The placental thyroid hormone synthesis pathway may be a target of maternal stress and modulate fetal programming of health and disease of offspring in a sex-specific fashion.

4.
Neurobiol Dis ; 169: 105720, 2022 07.
Article in English | MEDLINE | ID: mdl-35417751

ABSTRACT

BACKGROUND: Motor symptoms of Parkinson's disease (PD) are apparent after a high proportion of dopamine neurons in the substantia nigra have degenerated. The vast majority of PD cases are sporadic, and the underlying pathobiological causes are poorly understood. Adults exhibit great variability in the numbers of nigral dopamine neurons, suggesting that factors during embryonic or early life regulate the development and physiology of dopaminergic neurons. Furthermore, exposure to infections and inflammation in utero has been shown to affect fetal brain development in models of schizophrenia and autism. Here, we utilize a mouse maternal infection model to examine how maternal herpesvirus infection impacts dopaminergic neuron-related gene and protein expression in the adult offspring. METHODS: Pregnant mice were injected with murine cytomegalovirus (MCMV), murine gamma herpes virus-68 (MHV68) or phosphate buffered saline (PBS) at embryonic day 8.5. Offspring were sacrificed at eight weeks of age and midbrains were processed for whole genome RNA sequencing, DNA methylation analysis, targeted protein expression and high-performance liquid chromatography for quantification of dopamine and its metabolites. RESULTS: The midbrain of adult offspring from MHV68 infected dams had significantly decreased expression of genes linked to dopamine neurons (Th, Lmx1b, and Foxa1) and increased Lrrk2, a gene involved in familial PD and PD risk that associates with neuroinflammation. Deconvolution analysis revealed that the proportion of dopamine neuron genes in the midbrain was reduced. There was an overall increase in DNA methylation in the midbrain of animals from MHV68-infected dams and pathway analyses indicated mitochondrial dysfunction, with reductions in genes associated with ATP synthesis, mitochondrial respiratory chain, and mitochondrial translation in the offspring of dams infected with MHV68. TIGAR (a negative regulator of mitophagy) and SDHA (mitochondrial complex II subunit) protein levels were increased, and the levels of 3,4-dihydroxyphenylacetic acid (DOPAC) in the striatum were increased in these offspring compared to offspring from uninfected control dams. No such changes were observed in the offspring of dams infected with MCMV. CONCLUSION: Our data suggest that maternal infection with Herpesviridae, specifically MHV68, can trigger changes in the development of the midbrain that impact dopamine neuron physiology in adulthood. Our work is of importance for the understanding of neuronal susceptibility underlying neurodegenerative disease, with particular relevance for PD.


Subject(s)
Herpesviridae Infections , Herpesviridae , Neurodegenerative Diseases , Parkinson Disease , Animals , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Female , Herpesviridae/metabolism , Herpesviridae Infections/metabolism , Mesencephalon/metabolism , Mice , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Pregnancy , Substantia Nigra/metabolism
5.
Placenta ; 91: 59-65, 2020 02.
Article in English | MEDLINE | ID: mdl-32174308

ABSTRACT

INTRODUCTION: Placental viral infections are associated with fetal inflammation and adverse pregnancy outcomes. However, there have been limited studies on how placental macrophages in the villous and adjacent fetal umbilical endothelial cells respond to a viral insult. This study aimed to evaluate the communication between Hofbauer cells (HBCs) and human umbilical vein endothelial cells (HUVECs) during a viral infection. METHODS: HBCs were either uninfected or infected with the γ-herpesvirus, MHV-68, and the conditioned medium (CM) collected. HUVECs were exposed to HBC CM and the levels of the pro-neutrophilic response markers: IL-8; E-selectin; intercellular adhesion molecule 1 (ICAM-1); and vascular adhesion molecule 1 (VCAM-1) measured by ELISA and qPCR. The role of HBC-derived IL-1ß was investigated using an IL-1ß blocking antibody (Ab) or IL-1 receptor antagonist (IL-1Ra). RESULTS: MHV-68 infection of HBCs induced a significant increase in IL-1ß secretion. CM from infected HBCs induced HUVEC expression of IL-8, E-selectin, VCAM-1, ICAM-1 mRNA, and secretion of IL-8. The HUVEC response to the CM of MHV-infected HBCs was inhibited by a neutralizing IL-1ß Ab and by IL-1Ra. DISCUSSION: Virally-induced HBC IL-1ß activates HUVECs to generate a pro-neutrophilic response. This novel cell-cell communication pathway may play an important role in the genesis of fetal inflammation associated with placental viral infection.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Placenta/metabolism , Female , Herpesviridae , Human Umbilical Vein Endothelial Cells/virology , Humans , Intercellular Adhesion Molecule-1/metabolism , Macrophages/virology , Placenta/virology , Pregnancy , Vascular Cell Adhesion Molecule-1/metabolism
6.
Int J Tryptophan Res ; 12: 1178646919840321, 2019.
Article in English | MEDLINE | ID: mdl-31007529

ABSTRACT

BACKGROUND: The kynurenine pathway enzymes, breaking down tryptophan, are abundant in placental tissue. These metabolites are involved in immunoregulatory mechanisms, although the role of this pathway in pre-eclampsia (PE) has only begun to be characterized. Here, we determined tryptophan and metabolite levels together with the expression of kynurenine pathway enzymes and inflammatory factors in placental tissue from women with and without PE. METHODS: Thirty-six placentas (18 PE and 18 controls) were analyzed for expression of kynurenine pathway enzymes indoleamine-2,3-dioxygenase (IDO1 and 2), tryptophan-2,3-dioxygenase (TDO), kynurenine-3-mono-oxygenase (KMO) and quinolinate phosphoribosyltransferase (QPRT) as well as interleukin (IL)-1ß, IL-6, and serum amyloid A (SAA). Tryptophan and kynurenine content were measured using high-pressure liquid chromatography and quinolinic acid was measured using gas chromatography-mass spectrometry. CONCLUSIONS: Tryptophan content was reduced in placentas from women with PE. There was an increased kynurenine/tryptophan ratio in placentas from women with PE but no significant change in downstream metabolites. We confirmed a reduction in IDO1 expression and found a compensatory increase in TDO expression in placentas from women with PE. SAA was reduced in PE placentas compared with controls. Our data show that tryptophan content and the inflammatory mediator SAA are both compromised in placentas from women with PE. Further studies on the role of tryptophan catabolism and mediators of inflammation in sustaining healthy immunobiological pathways in the placenta are warranted.

7.
Front Immunol ; 10: 3134, 2019.
Article in English | MEDLINE | ID: mdl-32038643

ABSTRACT

Allergic asthma is a chronic pulmonary disorder fundamentally linked to immune dysfunction. Since the immune system begins developing in utero, prenatal exposures can affect immune programming and increase risk for diseases such as allergic asthma. Chronic psychosocial stress during pregnancy is one such risk factor, having been associated with increased risk for atopic diseases including allergic asthma in children. To begin to define the underlying causes of the association between maternal stress and allergic airway inflammation in offspring, we developed a mouse model of chronic heightened stress hormone during pregnancy. Continuous oral administration of corticosterone (CORT) to pregnant mice throughout the second half of pregnancy resulted in an ~2-fold increase in circulating hormone in dams with no concomitant increase in fetal circulation, similar to the human condition. To determine how prolonged heightened stress hormone affected allergic immunity in offspring, we induced allergic asthma with house dust mite (HDM) and examined the airway immune response to allergen. Female mice responded to HDM more frequently and had a more robust immune cell response compared to their male counterparts, irrespective of maternal treatment. Male offspring from CORT-treated dams had a greater number of inflammatory cells in the lung in response to HDM compared to males from control dams, while maternal treatment did not affect immune cell numbers in females. Alternatively, maternal CORT caused enhanced goblet cell hyperplasia in female offspring following HDM, an effect that was not observed in male offspring. In summary, prenatal exposure to mild, prolonged heightened stress hormone had sexually dimorphic effects on allergic inflammation in airways of adult offspring.


Subject(s)
Asthma/etiology , Asthma/immunology , Corticosterone/adverse effects , Pregnancy/psychology , Prenatal Exposure Delayed Effects/immunology , Animals , Corticosterone/metabolism , Female , Humans , Male , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Pregnancy/metabolism , Prenatal Exposure Delayed Effects/etiology , Pyroglyphidae/immunology , Stress, Physiological , Stress, Psychological
8.
PLoS One ; 13(6): e0199566, 2018.
Article in English | MEDLINE | ID: mdl-29912991

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0188645.].

9.
Cell Mol Immunol ; 15(12): 1010-1026, 2018 12.
Article in English | MEDLINE | ID: mdl-29907882

ABSTRACT

Pregnancy is a unique immunologic and microbial condition that requires an adequate level of awareness to provide a fast and protective response against pathogens as well as to maintain a state of tolerance to paternal antigens. Dysregulation of inflammatory pathways in the placenta triggered by pathogens is one of the main factors responsible for pregnancy complications. Type I IFNs are key molecules modulating immune responses at the level of the placenta and are crucial for protection of the pregnancy via their antiviral and immune modulatory properties. In this study, we elucidate the mechanisms controlling the basal expression of IFNß and its negative feedback. Using in vitro and in vivo animal models, we found that TLR signaling maintains basal IFNß levels through the TLR4-MyD88-independent TBK/IRF3 signaling pathway. We describe the role of the TAM receptor Axl in the regulation of IFNß function in human and mouse trophoblast cells. The absence of TAM receptors in vivo is associated with fetal demise due to dysregulation of IFNß expression and its pro-apoptotic downstream effectors. Collectively, our data describe a feedback signaling pathway controlling the expression and function of IFNß in the trophoblast that is essential for an effective response during viral and microbial infections.


Subject(s)
Interferon-beta/metabolism , Placenta/immunology , Trophoblasts/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Pregnancy Trimester, First , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Axl Receptor Tyrosine Kinase
10.
Am J Reprod Immunol ; 79(5): e12840, 2018 05.
Article in English | MEDLINE | ID: mdl-29520885

ABSTRACT

PROBLEM: Autism spectrum disorder (ASD) is one of the most commonly diagnosed neurodevelopmental disorders in the United States. While ASD can be significantly influenced by genetics, prenatal exposure to maternal infections has also been implicated in conferring risk. Despite this, the effects of several important maternal pathogens, such as cytomegalovirus (CMV) and herpes simplex virus 2 (HSV2), remain unknown. METHOD OF STUDY: We tested whether maternal CMV and/or HSV2 sero-positivity was associated with ASD symptoms in children. ELISA was used to assay for CMV IgG and HSV2 IgG in serum from the mothers of 82 children whose ASD symptoms were assessed at 3-6 years of age using the Social Responsiveness Scale version 2 (SRS-2). RESULTS: Associations between maternal viral serostatus and SRS-2 scores were estimated using linear regression with covariate adjustments. The children of mothers sero-positive for CMV, but not for HSV2, had SRS-2 scores 3.6-4.2 points higher, depending on the adjustment model, than sero-negative women, a significant finding, robust to several statistical adjustments. CONCLUSION: Our results suggest that maternal CMV infections may influence ASD symptoms. These findings are being further evaluated in ongoing prospective studies with larger population samples.


Subject(s)
Autistic Disorder/etiology , Autistic Disorder/virology , Cytomegalovirus Infections/complications , Cytomegalovirus/immunology , Autistic Disorder/immunology , Child , Child, Preschool , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Female , Herpes Genitalis/complications , Herpes Genitalis/immunology , Herpes Genitalis/virology , Herpesvirus 2, Human/immunology , Humans , Immunoglobulin G/blood , Mothers , Pregnancy
11.
PLoS One ; 12(11): e0188645, 2017.
Article in English | MEDLINE | ID: mdl-29190738

ABSTRACT

Preterm birth (PTB), or birth before 37 weeks gestation, is the leading cause of neonatal mortality worldwide. Cervical viral infections have been established as risk factors for PTB in women, although the mechanism leading to increased risk is unknown. Using a mouse model of pregnancy, we determined that intra-vaginal HSV2 infection caused increased rates of preterm birth following an intra-vaginal bacterial infection. HSV2 infection resulted in histological changes in the cervix mimicking cervical ripening, including significant collagen remodeling and increased hyaluronic acid synthesis. Viral infection also caused aberrant expression of estrogen and progesterone receptor in the cervical epithelium. Further analysis using human ectocervical cells demonstrated a role for Src kinase in virus-mediated changes in estrogen receptor and hyaluronic acid expression. In conclusion, HSV2 affects proteins involved in tissue hormone responsiveness, causes significant changes reminiscent of premature cervical ripening, and increases risk of preterm birth. Studies such as this improve our chances of identifying clinical interventions in the future.


Subject(s)
Cervical Length Measurement , Herpes Genitalis/pathology , Herpesvirus 2, Human/pathogenicity , Premature Birth , Animals , Escherichia coli Infections/complications , Escherichia coli Infections/physiopathology , Female , Herpes Genitalis/complications , Herpes Genitalis/physiopathology , Humans , Mice , Mice, Inbred C57BL , Models, Animal , Pregnancy
12.
Am J Reprod Immunol ; 78(5)2017 Nov.
Article in English | MEDLINE | ID: mdl-28846166

ABSTRACT

PROBLEM: Heightened maternal stress affects trophoblast function and increases risk for adverse pregnancy outcomes. METHODS OF STUDY: Studies were performed using the first-trimester trophoblast cell line, Sw.71. Cytokines were quantified using qPCR and ELISA. Epigenetic regulation of cytokines was characterized by inhibiting histone deacetylation (1 µmol/L suberoylanilide hydroxamic acid [SAHA]) or methylation (5 µmol/L 5-azacytidine), or with chromatin immunoprecipitation (ChIP) with a pan-acetyl histone-3 antibody. Invasion assays used Matrigel chambers. RESULTS: Cortisol inhibited expression of CSF2 (GM-CSF) and CSF3 (G-CSF) in trophoblast cells. Cortisol-associated inhibition was dependent on DNA methylation and was not affected by acetylation. There was also a modest decrease in trophoblast invasion, not dependent on loss of CSFs. CONCLUSION: In first-trimester trophoblast cells, the physiological glucocorticoid, cortisol, inhibited two cytokines with roles in placental development and decreased trophoblast invasion. Cortisol-associated changes in trophoblast function could increase the risk for immune-mediated abortion or other adverse pregnancy outcomes.


Subject(s)
Abortion, Spontaneous/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hydrocortisone/immunology , Stress, Psychological/immunology , Trophoblasts/immunology , Cell Line , DNA Methylation , Epigenesis, Genetic , Female , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Placentation , Pregnancy , Pregnancy Outcome , Pregnancy Trimester, First
13.
Trends Mol Med ; 23(9): 778-785, 2017 09.
Article in English | MEDLINE | ID: mdl-28803703

ABSTRACT

The 2015 Zika virus (ZIKV) outbreak caused global concern when it was determined to cause microcephaly, hearing loss, and other neurodevelopmental manifestations upon fetal exposure. Significant progress has been made in our understanding of the interactions between ZIKV and the pregnant host, but there is still a critical need to understand how ZIKV and other neurotropic viruses affect fetal neurodevelopment. Diaphanous-related formins (Diaphs) have recently been identified as microcephaly-associated proteins in humans and mice. Mutations in Diaphs affect the function of neural progenitor cells, much like prenatal viral infection. We present a novel hypothesis that viruses 'hijack' Diaphs in neural progenitor cells, causing autonomous differentiation and apoptosis of neural progenitor cells, which could potentially contribute to virus-associated neurological pathologies.


Subject(s)
Adaptor Proteins, Signal Transducing , Disease Outbreaks , Microcephaly , Neural Stem Cells , Zika Virus Infection , Zika Virus , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Formins , Humans , Microcephaly/epidemiology , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Zika Virus/genetics , Zika Virus/metabolism , Zika Virus Infection/epidemiology , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
14.
J Clin Invest ; 127(5): 1591-1599, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28459427

ABSTRACT

Despite the prevalence of viral infections in the American population, we still have a limited understanding of how they affect pregnancy and fetal development. Viruses can gain access to the decidua and placenta by ascending from the lower reproductive tract or via hematogenous transmission. Viral tropism for the decidua and placenta is then dependent on viral entry receptor expression in these tissues as well as on the maternal immune response to the virus. These factors vary by cell type and gestational age and can be affected by changes to the in utero environment and maternal immunity. Some viruses can directly infect the fetus at specific times during gestation, while some only infect the placenta. Both scenarios can result in severe birth defects or pregnancy loss. Systemic maternal viral infections can also affect the pregnancy, and these can be especially dangerous, because pregnant women suffer higher virus-associated morbidity and mortality than do nonpregnant counterparts. In this Review, we discuss the potential contributions of maternal, placental, and fetal viral infection to pregnancy outcome, fetal development, and maternal well-being.


Subject(s)
Fetal Diseases/mortality , Infectious Disease Transmission, Vertical , Placenta/virology , Pregnancy Complications, Infectious/mortality , Virus Diseases , Female , Humans , Placenta/pathology , Pregnancy , Risk Factors , Virus Diseases/mortality , Virus Diseases/transmission
15.
J Immunol ; 198(8): 3029-3032, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28264970

ABSTRACT

Pregnant women have greater mortality and complications associated with viral infections compared with the general population, but the reason for the increased susceptibility is not well defined. Placenta type I IFN is an important immune modulator and protects the pregnancy. We hypothesized that loss of placental IFN affects the regulation of the maternal immune system, resulting in the differential response to infections observed in pregnancy. Pregnant mice lacking the IFN-α/ß receptor (IFNAR) became viremic and had higher mortality compared with nonpregnant animals. Notably, an embryo with functional IFN signaling alone was sufficient to rescue the pregnant IFNAR-/- dam from virus-associated demise. Placental IFN was also an important regulator of viral replication in placental tissue and significantly affected viral transmission to the fetus. These findings highlight the role of fetal/placental IFN in the modulation of viral infection in the mother and fetus.


Subject(s)
Fetus/immunology , Herpesviridae Infections/immunology , Interferon Type I/immunology , Placenta/immunology , Pregnancy Complications, Infectious/immunology , Animals , Female , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Viral Load
16.
Am J Reprod Immunol ; 75(4): 451-60, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26892235

ABSTRACT

PROBLEM: Preterm birth (PTB) affects approximately 12% of pregnancies and at least 50% of cases have no known risk factors. We hypothesize that subclinical viral infections of the placenta are a factor sensitizing women to intrauterine bacterial infection. Specifically, we propose that viral-induced placental IFN-ß inhibition results in a robust inflammatory response to low concentrations of bacteria. METHODS: Human trophoblast SW.71, C57BL/6, and interferon (IFN) receptor knockout animals were used to determine IFN function. Illumina and Bio-Rad microarrays identified pathways. RESULTS: Inhibiting the IFN-ß pathway resulted in a significant increase in inflammatory cytokines such as IL-1B in response to LPS. Twist was positively correlated with IFN-ß expression and STAT3 phosphorylation and overexpressing Twist reduced IL-1B. Treating IFNAR-/- mice with low-dose LPS at E15.5 caused preterm birth. CONCLUSION: IFN-ß was identified as a key modulator of placental inflammation and, importantly, is commonly affected by viruses. We propose dysregulation of IFN-ß is a major determinant for preterm birth associated with polymicrobial infection.


Subject(s)
Interferon-beta/immunology , Pregnancy Complications, Infectious/immunology , Premature Birth/immunology , Receptor, Interferon alpha-beta/immunology , Trophoblasts/immunology , Virus Diseases/immunology , Animals , Cell Line, Transformed , Female , Gene Expression Regulation/immunology , Humans , Interferon-beta/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Pregnancy , Pregnancy Complications, Infectious/genetics , Pregnancy Complications, Infectious/pathology , Premature Birth/genetics , Premature Birth/microbiology , Premature Birth/virology , Receptor, Interferon alpha-beta/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Trophoblasts/microbiology , Trophoblasts/pathology , Trophoblasts/virology , Virus Diseases/genetics , Virus Diseases/pathology
17.
Am J Reprod Immunol ; 73(3): 199-213, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582523

ABSTRACT

Viral infections during pregnancy have long been considered benign conditions with a few notable exceptions, such as herpes virus. The recent Ebola outbreak and other viral epidemics and pandemics show how pregnant women suffer worse outcomes (such as preterm labor and adverse fetal outcomes) than the general population and non-pregnant women. New knowledge about the ways the maternal-fetal interface and placenta interact with the maternal immune system may explain these findings. Once thought to be 'immunosuppressed', the pregnant woman actually undergoes an immunological transformation, where the immune system is necessary to promote and support the pregnancy and growing fetus. When this protection is breached, as in a viral infection, this security is weakened and infection with other microorganisms can then propagate and lead to outcomes, such as preterm labor. In this manuscript, we review the major viral infections relevant to pregnancy and offer potential mechanisms for the associated adverse pregnancy outcomes.


Subject(s)
Pregnancy Complications, Infectious/immunology , Virus Diseases/immunology , Animals , Coinfection , Congenital Abnormalities/etiology , Female , Fetal Diseases/immunology , HIV Infections/congenital , HIV Infections/embryology , HIV Infections/immunology , HIV Infections/transmission , Hepatitis, Viral, Human/embryology , Hepatitis, Viral, Human/immunology , Hepatitis, Viral, Human/transmission , Herpesviridae Infections/embryology , Herpesviridae Infections/immunology , Herpesviridae Infections/transmission , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical , Influenza, Human/embryology , Influenza, Human/immunology , Maternal-Fetal Exchange/immunology , Obstetric Labor, Premature/etiology , Placenta/immunology , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/virology , Pregnancy Outcome , Risk , Rubella/embryology , Rubella/immunology , Rubella/transmission , Virus Diseases/transmission
18.
Am J Reprod Immunol ; 72(3): 270-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24995492

ABSTRACT

PROBLEM: During early pregnancy, macrophages and trophoblast come into close contact during placenta development, and regulated cross talk between these cellular compartments is crucial for maintaining a healthy pregnancy. As trophoblast cells constitutively secrete many chemokines and cytokines, we hypothesize that trophoblast-secreted factors can differentiate monocytes into a decidual phenotype. In this study, we describe a unique macrophage phenotype, following monocytes' exposure to trophoblast-soluble factors. METHOD OF STUDY: Peripheral blood monocytes were treated with or without conditioned media (CM) from first trimester trophoblast cells. Phenotypic changes and phagocytic capacity were determined by flow cytometry. Cytokine and chemokine production was determined by multiplex analysis. RESULTS: Monocytes exposed to trophoblast factors undergo morphologic changes characterized by a gain in size and complexity and acquire a unique phenotype characterized by gain of CD14 surface expression as well as CD16. The presence of CD14+/CD16+ macrophages was confirmed in normal decidua. These cells secrete higher levels of IL-1b, IL-10, and IP-10 and have increased capacity for phagocytosis. CONCLUSION: We demonstrate that trophoblast-secreted factors can induce monocyte differentiation into a unique macrophage phenotype. These findings suggest that the microenvironment of the placenta can modulate the phenotype of macrophages present at the decidua.


Subject(s)
Macrophages/immunology , Monocytes/cytology , Trophoblasts/immunology , Cell Differentiation , Cell Line , Cells, Cultured , Cytokines/immunology , GPI-Linked Proteins/immunology , Humans , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/immunology , Phagocytosis , Receptors, IgG/immunology , Transforming Growth Factor beta/pharmacology
19.
Am J Reprod Immunol ; 72(2): 107-16, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24995526

ABSTRACT

Progress in our understanding of the role of the maternal immune system during healthy pregnancy will help us better understand the role of the immune system in adverse pregnancy outcomes. In this review, we discuss our present understanding of the 'immunity of pregnancy' in the context of the response to cervical and placental infections and how these responses affect both the mother and the fetus. We discuss novel and challenging concepts that help explain the immunological aspects of pregnancy and how the mother and fetus respond to infection.


Subject(s)
Bacterial Infections/immunology , Fetus/immunology , Immune System , Placenta/immunology , Virus Diseases/immunology , Bacterial Infections/microbiology , Female , Fetus/microbiology , Fetus/virology , Humans , Immune Tolerance , Maternal-Fetal Exchange/immunology , Obstetric Labor, Premature/immunology , Obstetric Labor, Premature/microbiology , Obstetric Labor, Premature/virology , Placenta/microbiology , Placenta/virology , Pregnancy , Virus Diseases/microbiology
20.
Am J Reprod Immunol ; 72(2): 141-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24809430

ABSTRACT

Approximately half of all human embryo implantations result in failed pregnancy. Multiple factors may contribute to this failure, including genetic or metabolic abnormalities of the embryo. However, many of these spontaneous early abortion cases are attributed to poor uterine receptivity. Furthermore, although many fertility disorders have been overcome by a variety of assisted reproductive techniques, implantation remains the rate-limiting step for the success of the in vitro fertilization (IVF) treatments. We, as well as others, have demonstrated that endometrial biopsies performed either during the spontaneous, preceding cycle, or during the IVF cycle itself, significantly improve the rate of implantation, clinical pregnancies, and live births. These observations suggest that mechanical injury of the endometrium may enhance uterine receptivity by provoking the immune system to generate an inflammatory reaction. In strong support of this idea, we recently found that dendritic cells (DCs), an important cellular component of the innate immune system, play a critical role in successful implantation in a mouse model. In this review, we discuss the hypothesis that the injury-derived inflammation in the biopsy-treated patients generates a focus for uterine DCs and Mac accumulation that, in turn, enhance the endometrial expression of essential molecules that facilitate the interaction between the embryo and the uterine epithelium.


Subject(s)
Dendritic Cells/immunology , Embryo Implantation/immunology , Embryo Loss/immunology , Embryo, Mammalian/immunology , Macrophages/immunology , Biopsy , Dendritic Cells/cytology , Embryo Implantation/genetics , Embryo Loss/pathology , Embryo Loss/prevention & control , Embryo Transfer/methods , Embryo, Mammalian/metabolism , Endometrium/immunology , Female , Fertilization in Vitro , Humans , Inflammation/immunology , Macrophages/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...