Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Immunol ; 203(2): 500-510, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31167775

RESUMO

Preterm birth is a major contributor to neonatal mortality and morbidity, and infection is a major risk factor. Chorioamnionitis, inflammation of the placenta, and fetal membranes (FMs) are commonly observed in preterm birth and are characterized by neutrophil infiltration. However, interactions between FMs and neutrophils remain incompletely understood. The objectives of this study were to determine how FMs, with or without bacterial LPS stimulation, affect neutrophil recruitment, activation, and the formation of neutrophil extracellular traps (NETs) and to elucidate the signaling mechanisms involved. Using a combination of in vitro, ex vivo, and in vivo approaches, we show that human resting FMs can directly recruit neutrophils and induce them to produce proinflammatory factors. Furthermore, neutrophils release vital NETs in response to FM-derived factors. LPS-stimulated FMs further augmented neutrophil recruitment, inflammatory cytokine/chemokine secretion, and vital NET release and also induced reactive oxygen species production and degranulation. We demonstrate a role for FM-derived TNF-α in mediating these effects through activation of neutrophil p38 MAPK. We propose that, during infection, neutrophil recruitment and activation may neutralize pathogens, vital NET formation, and prolonged neutrophil viability, and in combination with degranulation, reactive oxygen species production and inflammatory chemokine/cytokine production may contribute to tissue injury at the maternal/fetal interface.


Assuntos
Armadilhas Extracelulares/imunologia , Membranas Extraembrionárias/imunologia , Lipopolissacarídeos/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Placenta/imunologia , Animais , Corioamnionite/imunologia , Citocinas/imunologia , Feminino , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/imunologia , Gravidez , Espécies Reativas de Oxigênio/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
2.
J Immunol ; 199(8): 2885-2895, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916522

RESUMO

Chorioamnionitis, premature rupture of fetal membranes (FMs), and subsequent preterm birth are associated with local infection and inflammation, particularly IL-1ß production. Although bacterial infections are commonly identified, other microorganisms may play a role in the pathogenesis. Because viral pandemics, such as influenza, Ebola, and Zika, are becoming more common, and pregnant women are at increased risk for associated complications, this study evaluated the impact that viral infection had on human FM innate immune responses. This study shows that a herpes viral infection of FMs sensitizes the tissue to low levels of bacterial LPS, giving rise to an exaggerated IL-1ß response. Using an ex vivo human FM explant system and an in vivo mouse model of pregnancy, we report that the mechanism by which this aggravated inflammation arises is through the inhibition of the TAM receptor, MERTK, and activation of the inflammasome. The TAM receptor ligand, growth arrest specific 6, re-establishes the normal FM response to LPS by restoring and augmenting TAM receptor and ligand expression, as well as by preventing the exacerbated IL-1ß processing and secretion. These findings indicate a novel mechanism by which viruses alter normal FM immune responses to bacteria, potentially giving rise to adverse pregnancy outcomes.


Assuntos
Membranas Extraembrionárias/imunologia , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 2/imunologia , Inflamassomos/metabolismo , Nascimento Prematuro/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Células Cultivadas , Corioamnionite , Feminino , Infecções por Herpesviridae/complicações , Humanos , Imunização , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Nascimento Prematuro/etiologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , c-Mer Tirosina Quinase
3.
Biol Reprod ; 92(1): 17, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25429091

RESUMO

Interest is growing in the role of viral infections and their association with adverse pregnancy outcomes. The trophoblast is permissive to viruses, but little is known about their impact on the placenta. We previously established that viral single stranded RNA (ssRNA), a Toll-like receptor 8 (TLR8) agonist, induces a restricted trophoblast pro-inflammatory cytokine/chemokine response by upregulating the secretion of interleukin (IL)-6 and IL-8. In parallel, the type I interferon, IFNbeta, is produced and acts back on the cell in an autocrine/paracrine manner to trigger caspase-3-dependent apoptosis. In this study, we sought to extend these findings by determining the mechanisms involved, examining whether viral ssRNA could induce a trophoblast antiviral response, and evaluating the influence of viral ssRNA on pregnancy outcome using a mouse model. Viral ssRNA induced human first-trimester trophoblast inflammation, type I interferon production, an antiviral response, and apoptosis in both a TLR8/MyD88-dependent and -independent manner. Furthermore, administration of viral ssRNA to pregnant mice induced placental caspase-3 activation, a pro-inflammatory cytokine/chemokine, type I interferon, and antiviral response as well as immune cell infiltration. Thus, ssRNA viral infections may compromise pregnancy by altering placental trophoblast survival and function through both TLR8 and non-TLR8 signaling pathways, leading to immune changes at the maternal-fetal interface.


Assuntos
Imunidade Inata , Inflamação , RNA Viral/fisiologia , Receptor 8 Toll-Like/fisiologia , Trofoblastos/imunologia , Animais , Caspase 3/metabolismo , Células Cultivadas , Feminino , Humanos , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos ICR , Fator 88 de Diferenciação Mieloide/fisiologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Trofoblastos/virologia
4.
Biol Reprod ; 90(2): 39, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24429216

RESUMO

Bacterial infection-associated inflammation is thought to be a major cause of preterm premature rupture of membranes. Proinflammatory cytokines, such as interleukin 1B (IL1B), can weaken fetal membranes (FM) by upregulating matrix metalloproteinases and inducing apoptosis. The mechanism by which infection leads to inflammation at the maternal-fetal interface and subsequent preterm birth is thought to involve innate immune pattern recognition receptors (PRR), such as the Toll-like receptors (TLR) and Nod-like receptors (NLR), which recognize pathogen-associated molecular patterns (PAMPs). The objective of this study was to determine the cytokine profile generated by FMs in response to the bacterial TLR and NLR agonists peptidoglycan (PDG; TLR2), lipopolysaccharide (LPS; TLR4), flagellin (TLR5), CpG ODN (TLR9), iE-DAP (Nod1), and MDP (Nod2). PDG, LPS, flagellin, iE-DAP, and MDP triggered FMs to generate an inflammatory response, but the cytokine profiles were distinct for each TLR and NLR agonist, and only IL1B and RANTES were commonly upregulated in response to all five PAMPs. CpG ODN, in contrast, had a mild stimulatory effect only on MCP-1 and primarily downregulated basal FM cytokine production. IL1B secretion induced by PDG, LPS, flagellin, iE-DAP, and MDP was associated with its processing. Furthermore, FM IL1B secretion in response to TLR2, TLR4, and TLR5 activation was caspase 1-dependent, whereas Nod1 and Nod2 induced IL1B secretion independent of caspase 1. These findings demonstrate that FMs respond to different bacterial TLR and NLR PAMPs by generating distinct inflammatory cytokine profiles through distinct mechanisms that are specific to the innate immune PRR activated.


Assuntos
Citocinas/metabolismo , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Flagelina/farmacologia , Lipopolissacarídeos/farmacologia , Proteínas Adaptadoras de Sinalização NOD/agonistas , Receptores Toll-Like/agonistas , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacologia , Membranas Extraembrionárias/imunologia , Feminino , Humanos , Interleucina-1beta/metabolismo , Proteínas Adaptadoras de Sinalização NOD/genética , Proteínas Adaptadoras de Sinalização NOD/metabolismo , Proteína Adaptadora de Sinalização NOD2/farmacologia , Oligodesoxirribonucleotídeos/farmacologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
5.
Hum Reprod Update ; 29(5): 675-693, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37290428

RESUMO

BACKGROUND: The endometrium is a highly dynamic tissue that undergoes dramatic proliferation and differentiation monthly in order to prepare the uterus for implantation and pregnancy. Intrauterine infection and inflammation are being increasingly recognized as potential causes of implantation failure and miscarriage, as well as obstetric complications later in gestation. However, the mechanisms by which the cells of the endometrium respond to infection remain understudied and recent progress is slowed in part owing to similar overlapping studies being performed in different species. OBJECTIVE AND RATIONALE: The aim of this scoping review is to systematically summarize all published studies in humans and laboratory animals that have investigated the innate immune sensing and response of the endometrium to bacteria and viruses, and the signaling mechanisms involved. This will enable gaps in our knowledge to be identified to inform future studies. SEARCH METHODS: The Cochrane Library, Ovid Embase/Medline, PubMed, Scopus, Google Scholar, and Web of Science databases were searched using a combination of controlled and free text terms for uterus/endometrium, infections, and fertility to March 2022. All primary research papers that have reported on endometrial responses to bacterial and viral infections in the context of reproduction were included. To focus the scope of the current review, studies in domesticated animals, included bovine, porcine, caprine, feline, and canine species were excluded. OUTCOMES: This search identified 42 728 studies for screening and 766 full-text studies were assessed for eligibility. Data was extracted from 76 studies. The majority of studies focused on endometrial responses to Escherichia coli and Chlamydia trachomatis, with some studies of Neisseria gonorrhea, Staphylococcus aureus, and the Streptococcus family. Endometrial responses have only been studied in response to three groups of viruses thus far: HIV, Zika virus, and the herpesvirus family. For most infections, both cellular and animal models have been utilized in vitro and in vivo, focusing on endometrial production of cytokines, chemokines, and antiviral/antimicrobial factors, and the expression of innate immune signaling pathway mediators after infection. This review has identified gaps for future research in the field as well as highlighted some recent developments in organoid systems and immune cell co-cultures that offer new avenues for studying endometrial responses to infection in more physiologically relevant models that could accelerate future findings in this area. WIDER IMPLICATIONS: This scoping review provides an overarching summary and benchmark of the current state of research on endometrial innate immune responses to bacterial and viral infection. This review also highlights some exciting recent developments that enable future studies to be designed to deepen our understanding of the mechanisms utilized by the endometrium to respond to infection and their downstream effects on uterine function.


Assuntos
Viroses , Infecção por Zika virus , Zika virus , Gravidez , Feminino , Animais , Bovinos , Gatos , Cães , Humanos , Suínos , Cabras , Endométrio/metabolismo , Útero/metabolismo , Bactérias , Viroses/metabolismo
6.
J Reprod Immunol ; 140: 103126, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32289593

RESUMO

The maternal-fetal interface possesses innate immune strategies to protect against infections. We previously reported that prior viral infection of human fetal membranes (FMs) in vitro and mouse FMs in vivo sensitized the tissue to low dose bacterial LPS leading to augmented inflammation. The objective of this study was to examine FM production of type I interferons (IFNs) and IFN-stimulated genes (ISGs) in the context of this polymicrobial model. Human FM explants and pregnant C57BL/6 mice were treated with or without low dose LPS following exposure to media or the γ-herpes virus, MHV-68. FM RNA was analyzed by qRT-PCR for type I IFNs, ISGs, upstream signaling, and MHV-68 open reading frames (ORFs). Pre-exposure to MHV-68 followed by LPS treatment inhibited the ability of LPS to induce human FM type I IFNs (IFNA, IFNB); ISGs (OAS, MxA, APOBEC3G) and upstream signaling mediators (RIG-I, TBK-1). Signaling mediators IRF-3 and IRF-7 were also reduced. In mouse FMs, pre-exposure to MHV-68 followed by LPS treatment reduced the ability of LPS to upregulate Ifna, Ifnb, Mxa, Irf7, and also reduced Irf3. MHV-68 infection of FMs induced ORF45 which targets IRF-7, and this was further augmented in response to a combination of MHV-68 and LPS. Together, these findings indicate that a viral infection blunts FM type I IFN production and signaling in response to LPS leading to a suppressed ISG response. Our studies suggest that a viral infection inhibits this protective FM response by negatively regulating IRF-7 through ORF45, leaving the maternal-fetal interface vulnerable to further viral attack.


Assuntos
Membranas Extraembrionárias/metabolismo , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/imunologia , Interferon Tipo I/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Proteínas Imediatamente Precoces/genética , Tolerância Imunológica , Inflamação , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/genética , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
7.
Am J Reprod Immunol ; 80(5): e13044, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30175447

RESUMO

PROBLEM: Hyperglycemia increases the risk of preeclampsia. Hyperglycemia induces a placental trophoblast inflammatory (IL-1ß, IL-6, IL-8), antiangiogenic (sFlt-1, sEndoglin), and anti-migratory profile. The IL-1ß response is mediated via inflammasome activation by the damage-associated molecular pattern (DAMP), uric acid. The objective of this study was to determine the role of high-mobility group box-1 (HMGB1), a DAMP that activates Toll-like receptor 4 (TLR4), in human first trimester trophoblast responses to hyperglycemia. The trophoblast response to excess glucose under different oxygen tensions was also investigated. METHOD OF STUDY: The human first trimester trophoblast cell line (Sw.71) was exposed to glucose mimicking normoglycemia (5 mmol/L) and hyperglycemia (10 mmol/L), either alone or with the TLR4 antagonist, LPS-RS; or the HMGB1 inhibitor, glycyrrhizin. Cells were also treated with glucose under hyperoxic (21% O2 ), normoxic (8% O2 ), and hypoxic (2% O2 ) conditions. Cell-free supernatants were assayed by ELISA and bioassays for inflammatory: IL-1ß, IL-6, and IL-8; inflammasome-associated: uric acid and caspase-1; angiogenic: sEndoglin, sFlt-1, and PlGF; and the DAMP, HMGB1. Cell migration was measured using a two-chamber colormetric assay. RESULTS: Excess glucose triggered a trophoblast sterile inflammatory IL-8 and antimigratory response through HMGB1 activation of TLR4. The IL-1ß and sFlt-1/PlGF response was TLR4-mediated, but HMGB1-independent, suggesting another DAMP may be involved. Hyperoxia rather than normoxia or hypoxia was a major driver of trophoblast dysfunction in response to excess glucose. CONCLUSION: The findings from this study indicate a novel mechanism by which hyperglycemia may impact trophoblast function, early placentation, and ultimately, pregnancy outcome.


Assuntos
Alarminas/metabolismo , Proteína HMGB1/metabolismo , Hiperglicemia/imunologia , Inflamação/imunologia , Pré-Eclâmpsia/imunologia , Receptor 4 Toll-Like/metabolismo , Trofoblastos/imunologia , Linhagem Celular , Movimento Celular , Feminino , Ácido Glicirrízico/farmacologia , Proteína HMGB1/antagonistas & inibidores , Humanos , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Gravidez , Risco , Transdução de Sinais , Trofoblastos/patologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Am J Reprod Immunol ; 80(1): e12861, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29709093

RESUMO

PROBLEM: Chorioamnionitis and infection-associated inflammation are major causes of preterm birth. Magnesium sulfate (MgSO4 ) is widely used in obstetrics as a tocolytic; however, its mechanism of action is unclear. This study sought to investigate how MgSO4 modulates infection-associated inflammation in fetal membranes (FMs), and whether the response was time dependent. METHOD OF STUDY: Human FM explants were treated with or without bacterial lipopolysaccharide (LPS); with or without MgSO4 added either: 1 hour before LPS; at the same time as LPS; 1 hour post-LPS; or 2 hours post-LPS. Explants were also treated with or without viral dsRNA and LPS, alone or in combination; and MgSO4 added 1 hour post-LPS After 24 hours, supernatants were measured for cytokines/chemokines; and tissue lysates measured for caspase-1 activity. RESULTS: Lipopolysaccharide-induced FM inflammation by upregulating the secretion of a number of inflammatory cytokines/chemokines. Magnesium sulfate administered 1-hour post-LPS inhibited FM secretion of IL-1ß, IL-6, G-CSF, RANTES, and TNFα. Magnesium sulfate administered 2 hours post-LPS augmented FM secretion of these factors as well as IL-8, IFNγ, VEGF, GROα and IP-10. Magnesium sulfate delivered 1- hour post-LPS inhibited LPS-induced caspase-1 activity, and inhibited the augmented IL-1ß response triggered by combination viral dsRNA and LPS. CONCLUSION: Magnesium sulfate differentially modulates LPS-induced FM inflammation in a time-dependent manner, in part through its modulation of caspase-1 activity. Thus, the timing of MgSO4 administration may be critical in optimizing its anti-inflammatory effects in the clinical setting. MgSO4 might also be useful at preventing FM inflammation triggered by a polymicrobial viral-bacterial infection.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Inflamação/tratamento farmacológico , Sulfato de Magnésio/farmacologia , Caspase 1/metabolismo , Corioamnionite/metabolismo , Citocinas/metabolismo , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Gravidez , Nascimento Prematuro/prevenção & controle , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
9.
Arthritis Rheumatol ; 70(6): 891-902, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29342502

RESUMO

OBJECTIVE: Women with antiphospholipid antibodies (aPL) are at risk for pregnancy complications associated with poor placentation and placental inflammation. Although these antibodies are heterogeneous, some anti-ß2 -glycoprotein I (anti-ß2 GPI) antibodies can activate Toll-like receptor 4 (TLR-4) and NLRP3 in human first-trimester trophoblasts. The objective of this study was to determine the role of negative regulators of TLR and inflammasome function in aPL-induced trophoblast inflammation. METHODS: Human trophoblasts were not treated or were treated with anti-ß2 GPI aPL or control IgG in the presence or absence of the common TAM (TYRO3, AXL, and Mer tyrosine kinase [MERTK]) receptor ligand growth arrest-specific protein 6 (GAS6) or the autophagy-inducer rapamycin. The expression and function of the TAM receptor pathway and autophagy were measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). Antiphospholipid antibody-induced trophoblast inflammation was measured by qRT-PCR, activity assays, and ELISA. RESULTS: Anti-ß2 GPI aPL inhibited trophoblast TAM receptor function by reducing cellular expression of the receptor tyrosine kinases AXL and MERTK and the ligand GAS6. The addition of GAS6 blocked the effects of aPL on the TLR-4-mediated interleukin-8 (IL-8) response. However, the NLRP3 inflammasome-mediated IL-1ß response was not affected by GAS6, suggesting that another regulatory pathway was involved. Indeed, anti-ß2 GPI aPL inhibited basal trophoblast autophagy, and reversing this with rapamycin inhibited aPL-induced inflammasome function and IL-1ß secretion. CONCLUSION: Basal TAM receptor function and autophagy may serve to inhibit trophoblast TLR and inflammasome function, respectively. Impairment of TAM receptor signaling and autophagy by anti-ß2 GPI aPL may allow subsequent TLR and inflammasome activity, leading to a robust inflammatory response.


Assuntos
Anticorpos Antifosfolipídeos/imunologia , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Receptor 4 Toll-Like/imunologia , Trofoblastos/imunologia , Linhagem Celular , Feminino , Humanos , Imunossupressores/administração & dosagem , Inflamação/induzido quimicamente , Gravidez , Primeiro Trimestre da Gravidez/imunologia , Sirolimo/administração & dosagem
10.
Am J Reprod Immunol ; 76(1): 29-37, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27108773

RESUMO

PROBLEM: Risk factors for preterm birth include placental abruption, giving rise to excessive decidual thrombin, and intrauterine bacterial infection. Human endometrial endothelial cells (HEECs) express Toll-like receptors (TLRs), and infection-derived agonists trigger HEECs to generate specific inflammatory responses. As thrombin, in addition to inducing coagulation, can contribute to inflammation, its effect on HEEC inflammatory responses to the TLR4 agonist, bacterial lipopolysaccharide (LPS), was investigated. METHOD OF STUDY: HEECs were pre-treated with or without thrombin or specific protease-activated receptor (PAR) agonists, followed by treatment with or without LPS. Supernatants were measured for cytokines and chemokines by ELISA and multiplex analysis. RESULTS: Thrombin significantly and synergistically augmented LPS-induced HEEC secretion of interleukin (IL)-6, IL-8, granulocyte colony-stimulating factor (G-CSF), and growth-regulated oncogene-alpha (GRO-α), and significantly augmented monocyte chemotactic protein (MCP)-1, tumor necrosis factor-alpha (TNF-α), and vascular endothelial growth factor (VEGF) secretion additively. Similar to thrombin, a PAR1 agonist synergistically augmented the LPS-induced HEEC secretion of inflammatory IL-6, IL-8, G-CSF, and GRO-α. CONCLUSION: Thrombin, via PAR1 activation, synergistically augments LPS-induced HEEC production of chemokines involved in immune cell recruitment and survival, suggesting a mechanism by which intrauterine abruption and bacterial infection may together be associated with an aggravated uterine inflammatory response.


Assuntos
Endométrio/imunologia , Células Endoteliais/imunologia , Lipopolissacarídeos/toxicidade , Receptor PAR-1/imunologia , Trombina/imunologia , Citocinas/imunologia , Feminino , Humanos
11.
Am J Reprod Immunol ; 70(3): 190-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23621614

RESUMO

PROBLEM: Human endometrial endothelial cell (HEEC) innate immunity remains poorly characterized. Based on their direct contact with the circulation, HEECs are uniquely positioned to be exposed to viral infections. This study evaluated the innate immune response generated by HEECs after exposure to the TLR3 agonist, Poly(I:C) and the TLR8 agonist, viral ssRNA. METHOD OF STUDY: HEECs were treated with or without Poly(I:C) or ssRNA. Culture supernatants were measured for cytokines by multiplex analysis. RNA was analyzed by qRT-PCR for type I interferons and antiviral factors. RESULTS: Treatment of HEECs with Poly(I:C) rapidly upregulated the secretion of IL-2, IL-6, IL-8, IFN-γ, G-CSF, GM-CSF, MCP-1, MIP-1ß, RANTES, and GRO-α after 12 hr, while ssRNA treatment induced the slower secretion of IL-6, IL-8, IFN-γ, G-CSF, VEGF, and GRO-α after 24 hr. Both viral components induced HEEC IFN-α and IFN-ß expression. While treatment with Poly(I:C) induced APOBEC3G and OAS expression, treatment with ssRNA upregulated APOBEC3G and M×A mRNA. CONCLUSION: Our findings demonstrate that HEECs can differentially sense and respond to viral components by generating distinct inflammatory and antiviral immune responses, indicating that these cells likely play an active role in the immune protection of the uterus toward viral infections.


Assuntos
Antivirais/metabolismo , Endométrio/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Inflamação/imunologia , Poli I-C/farmacologia , RNA Viral/farmacologia , Receptor 3 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Células Cultivadas , Citocinas/metabolismo , Endométrio/citologia , Endométrio/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Poli I-C/genética , Poli I-C/imunologia , RNA Viral/genética , RNA Viral/imunologia , Receptor 3 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo
12.
PLoS One ; 8(10): e77249, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143215

RESUMO

While infection-induced placental inflammation is a common mechanism of adverse pregnancy outcome, some pathogens can also trigger placental apoptosis, and Toll-like receptors (TLRs) mediate this response. Treatment of human first trimester trophoblast cells with bacterial peptidoglycan (PDG) reduces their constitutive secretion of IL-6 protein and induces apoptosis. This apoptotic response is dependent upon the cell's expression of TLR1, TLR2 and TLR10, and their lack of TLR6, such that ectopic expression of TLR6 prevents PDG-induced apoptosis and restores IL-6 production. In this current study we have identified three microRNAs (miRs) that regulate TLR2-mediated responses in the human trophoblast. Herein we report that miR-329 plays a pivotal role in mediating PDG-induced trophoblast apoptosis and inhibition of IL-6 mRNA expression by targeting the NF-κB subunit, p65. TLR2 activation by PDG upregulates miR-329 expression and inhibits NF-κB p65 and IL-6 mRNA, and this is reversed by the presence of TLR6. Moreover, inhibition of miR-329 prevents PDG-induced inhibition of NF-κB p65 and IL-6 mRNA expression, and restores cell survival. In addition, we have found miR-23a and let-7c to directly regulate PDG-mediated inhibition of IL-6 mRNA. TLR2 activation by PDG upregulates miR23a and let-7c expression and this is reversed by the presence of TLR6. Furthermore, inhibition of both miR23a and let-7c prevents PDG-inhibition of trophoblast IL-6 mRNA expression. Together, our findings suggest that multiple miRs are involved in the molecular regulation of TLR2-mediated responses in the trophoblast towards gram-positive bacterial components.


Assuntos
Apoptose , Interleucina-6/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Receptor 2 Toll-Like/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo , Apoptose/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Peptidoglicano/farmacologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor 6 Toll-Like/metabolismo , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/metabolismo , Trofoblastos/efeitos dos fármacos
13.
Am J Reprod Immunol ; 69(1): 33-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22967004

RESUMO

PROBLEM: Preterm premature rupture of fetal membranes (pPROM) occurs in 30-40% of spontaneous preterm births (PTB) and is associated with intra-amniotic infection and inflammation. The membranes may sense and respond to microbes via Toll-like receptors (TLRs); however, little is known about their expression and regulation in this tissue. The objective of this study was to evaluate the expression of TLRs 1-10 in fetal membranes after exposure to pathogens associated with intra-amniotic infection and PTB. METHOD OF STUDY: Normal human term fetal membrane explants were exposed to various bacteria. After 24 hrs, RNA was extracted and quantitative RT-PCR performed for TLRs1-10. RESULTS: Treatment of fetal membranes with Mycoplasma hominis increased expression of TLR4, TLR6, and TLR8 mRNA. Ureaplasma parvum upregulated TLR8 mRNA, and Porphyromonas gingivalis significantly increased fetal membrane TLR7 expression. In contrast, treatment with Gram-negative Escherichia coli (and its cell wall component lipopolysaccharide) downregulated TLR10 mRNA. No effect was detected for Ureaplasma urealyticum, Gardnerella vaginalis, or Group B Streptococcus. CONCLUSION: These findings demonstrate that different types of bacteria have distinct effects on fetal membrane TLR expression patterns. Moreover, these findings highlight the disparity of fetal membrane responses to infection and thus suggest heterogeneity in the mechanisms by which infection-associated pregnancy complications, such as pPROM and PTB, arise.


Assuntos
Infecções Bacterianas/imunologia , Membranas Extraembrionárias/imunologia , Ruptura Prematura de Membranas Fetais/imunologia , Complicações Infecciosas na Gravidez/imunologia , Receptores Toll-Like/metabolismo , Infecções Bacterianas/microbiologia , Células Cultivadas , Escherichia coli/imunologia , Membranas Extraembrionárias/microbiologia , Feminino , Ruptura Prematura de Membranas Fetais/microbiologia , Gardnerella vaginalis/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Lipopolissacarídeos/imunologia , Mycoplasma hominis/imunologia , Técnicas de Cultura de Órgãos , Porphyromonas gingivalis/imunologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Nascimento Prematuro/etiologia , Nascimento Prematuro/prevenção & controle , Streptococcus/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Ureaplasma/imunologia , Ureaplasma urealyticum/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA