Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-35163786

RESUMO

Labor is a process of inflammation and hormonal changes involving both fetal and maternal compartments. MicroRNA-132-3p (miR-132-3p) has been reported to be involved in the development of inflammation-related diseases. However, little is known about its potential role in labor onset. This study aimed to explore the mechanism of miR-132-3p in amnion for labor initiation. In the mouse amnion membranes, the expression of miR-132-3p was found to increase gradually during late gestation. In human amniotic epithelial cell line (WISH), upregulation of miR-132-3p was found to increase proinflammatory cytokines and cyclooxygenase 2 (COX2) as well as prostaglandin E2 (PGE2), which was suppressed by miR-132-3p inhibitor. Dual-specificity phosphatase 9 (DUSP9) was identified as a novel target gene of miR-132-3p, which could be negatively regulated by miR-132-3p. DUSP9 was present in the mouse amnion epithelial cells, with a decrease in its abundance at 18.5 days post coitum (dpc) relative to 15.5 dpc. Silencing DUSP9 was found to facilitate the expression of proinflammatory cytokines and COX2 as well as PGE2 secretion in WISH cells, which could be attenuated by p38 inhibitor SB203580 or JNK inhibitor SP600125. Additionally, intraperitoneal injection of pregnant mice with miR-132-3p agomir not only caused preterm birth, but also promoted the abundance of COX2 as well as phosphorylated JNK and p38 levels, and decreased DUSP9 level in mouse amnion membranes. Collectively, miR-132-3p might participate in inflammation and PGE2 release via targeting DUSP9-dependent p38 and JNK signaling pathways to cause preterm birth.


Assuntos
Âmnio/imunologia , Fosfatases de Especificidade Dupla/genética , Inflamação/genética , Trabalho de Parto/genética , MicroRNAs/genética , Âmnio/citologia , Âmnio/metabolismo , Animais , Antracenos/farmacologia , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Gravidez , Piridinas/farmacologia
2.
Front Immunol ; 12: 735324, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745106

RESUMO

Intra-amniotic infection and inflammation (IAI) affect fetal development and are highly associated with preterm labor and premature rupture of membranes, which often lead to adverse neonatal outcomes. Human amniotic membrane (hAM), the inner part of the amnio-chorionic membrane, protects the embryo/fetus from environmental dangers, including microbial infection. However, weakened amnio-chorionic membrane may be breached or pathogens may enter through a different route, leading to IAI. The hAM and human amniotic fluid (hAF) respond by activation of all components of the innate immune system. This includes changes in 1) hAM structure, 2) presence of immune cells, 3) pattern recognition receptors, 4) cytokines, 5) antimicrobial peptides, 6) lipid derivatives, and 7) complement system. Herein we provide a comprehensive and integrative review of the current understanding of the innate immune response in the hAM and hAF, which will aid in design of novel studies that may lead to breakthroughs in how we perceive the IAI.


Assuntos
Âmnio/imunologia , Líquido Amniótico/imunologia , Bactérias/imunologia , Infecções Bacterianas/imunologia , Corioamnionite/imunologia , Imunidade Inata , Complicações Infecciosas na Gravidez/imunologia , Âmnio/metabolismo , Âmnio/microbiologia , Líquido Amniótico/metabolismo , Líquido Amniótico/microbiologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Corioamnionite/metabolismo , Corioamnionite/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Trabalho de Parto Prematuro/imunologia , Trabalho de Parto Prematuro/metabolismo , Trabalho de Parto Prematuro/microbiologia , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/microbiologia , Nascimento Prematuro , Transdução de Sinais
3.
Biomolecules ; 11(8)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34439812

RESUMO

Intra-amniotic infections (IAI) are one of the reasons for preterm birth. High mobility group box 1 (HMGB1) is a nuclear protein with various physiological functions, including tissue healing. Its excessive extracellular release potentiates inflammatory reaction and can revert its action from beneficial to detrimental. We infected the amniotic fluid of a pig on the 80th day of gestation with 1 × 104 colony forming units (CFUs) of E. coli O55 for 10 h, and evaluated the appearance of HMGB1, receptor for glycation endproducts (RAGE), and Toll-like receptor (TLR) 4 in the amniotic membrane and fluid. Sham-infected amniotic fluid served as a control. The expression and release of HMGB1 were evaluated by Real-Time PCR, immunofluorescence, immunohistochemistry, and ELISA. The infection downregulated HMGB1 mRNA expression in the amniotic membrane, changed the distribution of HMGB1 protein in the amniotic membrane, and increased its level in amniotic fluid. All RAGE mRNA, protein expression in the amniotic membrane, and soluble RAGE level in the amniotic fluid were downregulated. TLR4 mRNA and protein expression and soluble TLR4 were all upregulated. HMGB1 is a potential target for therapy to suppress the exaggerated inflammatory response. This controlled expression and release can, in some cases, prevent the preterm birth of vulnerable infants. Studies on suitable animal models can contribute to the development of appropriate therapy.


Assuntos
Infecções por Escherichia coli/veterinária , Escherichia coli/patogenicidade , Proteína HMGB1/genética , Complicações Infecciosas na Gravidez/veterinária , RNA Mensageiro/genética , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor 4 Toll-Like/genética , Âmnio/imunologia , Âmnio/microbiologia , Âmnio/patologia , Líquido Amniótico/imunologia , Líquido Amniótico/microbiologia , Animais , Modelos Animais de Doenças , Escherichia coli/crescimento & desenvolvimento , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Regulação da Expressão Gênica , Proteína HMGB1/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Gravidez , Complicações Infecciosas na Gravidez/genética , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/microbiologia , Nascimento Prematuro/prevenção & controle , RNA Mensageiro/imunologia , Receptor para Produtos Finais de Glicação Avançada/imunologia , Transdução de Sinais , Suínos , Receptor 4 Toll-Like/imunologia
4.
Int J Mol Sci ; 22(15)2021 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-34361111

RESUMO

Maternal smoking is a risk factor of preterm prelabor rupture of the fetal membranes (pPROM), which is responsible for 30% of preterm births worldwide. Cigarettes induce oxidative stress and inflammation, mechanisms both implicated in fetal membranes (FM) weakening. We hypothesized that the receptor for advanced glycation end-products (RAGE) and its ligands can result in cigarette-dependent inflammation. FM explants and amniotic epithelial cells (AECs) were treated with cigarette smoke condensate (CSC), combined or not with RAGE antagonist peptide (RAP), an inhibitor of RAGE. Cell suffering was evaluated by measuring lactate dehydrogenase (LDH) medium-release. Extracellular HMGB1 (a RAGE ligand) release by amnion and choriodecidua explants were checked by western blot. NF-κB pathway induction was determined by a luciferase gene reporter assay, and inflammation was evaluated by cytokine RT-qPCR and protein quantification. Gelatinase activity was assessed using a specific assay. CSC induced cell suffering and HMGB1 secretion only in the amnion, which is directly associated with a RAGE-dependent response. CSC also affected AECs by inducing inflammation (cytokine release and NFκB activation) and gelatinase activity through RAGE engagement, which was linked to an increase in extracellular matrix degradation. This RAGE dependent CSC-induced inflammation associated with an increase of gelatinase activity could explain a pathological FM weakening directly linked to pPROM.


Assuntos
Âmnio/patologia , Células Epiteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Fumaça/efeitos adversos , Adulto , Âmnio/efeitos dos fármacos , Âmnio/imunologia , Âmnio/metabolismo , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptor para Produtos Finais de Glicação Avançada
5.
PLoS One ; 16(6): e0253456, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34170941

RESUMO

BACKGROUND: Lung inflammation and impaired alveolarization are hallmarks of bronchopulmonary dysplasia (BPD). We hypothesize that human amnion epithelial cells (hAECs) are anti-inflammatory and reduce lung injury in preterm lambs born after antenatal exposure to inflammation. METHODS: Pregnant ewes received either intra-amniotic lipopolysaccharide (LPS, from E.coli 055:B5; 4mg) or saline (Sal) on day 126 of gestation. Lambs were delivered by cesarean section at 128 d gestation (term ~150 d). Lambs received intravenous hAECs (LPS/hAECs: n = 7; 30x106 cells) or equivalent volumes of saline (LPS/Sal, n = 10; or Sal/Sal, n = 9) immediately after birth. Respiratory support was gradually de-escalated, aimed at early weaning from mechanical ventilation towards unassisted respiration. Lung tissue was collected 1 week after birth. Lung morphology was assessed and mRNA levels for inflammatory mediators were measured. RESULTS: Respiratory support required by LPS/hAEC lambs was not different to Sal/Sal or LPS/Sal lambs. Lung tissue:airspace ratio was lower in the LPS/Sal compared to Sal/Sal lambs (P<0.05), but not LPS/hAEC lambs. LPS/hAEC lambs tended to have increased septation in their lungs versus LPS/Sal (P = 0.08). Expression of inflammatory cytokines was highest in LPS/hAECs lambs. CONCLUSIONS: Postnatal administration of a single dose of hAECs stimulates a pulmonary immune response without changing ventilator requirements in preterm lambs born after intrauterine inflammation.


Assuntos
Âmnio , Células Epiteliais , Lipopolissacarídeos/toxicidade , Pulmão , Pneumonia , Âmnio/imunologia , Âmnio/patologia , Animais , Animais Recém-Nascidos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/transplante , Feminino , Xenoenxertos , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/imunologia , Pulmão/patologia , Masculino , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/terapia , Ovinos
6.
J Innate Immun ; 13(2): 63-82, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33152737

RESUMO

Intra-amniotic infection, the invasion of microbes into the amniotic cavity resulting in inflammation, is a clinical condition that can lead to adverse pregnancy outcomes for the mother and fetus as well as severe long-term neonatal morbidities. Despite much research focused on the consequences of intra-amniotic infection, there remains little knowledge about the innate immune cells that respond to invading microbes. We performed RNA-seq of sorted amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection to determine the transcriptomic differences between these innate immune cells. Further, we sought to identify specific transcriptomic pathways that were significantly altered by the maternal or fetal origin of amniotic fluid neutrophils and monocytes/macrophages, the presence of a severe fetal inflammatory response, and pregnancy outcome (i.e., preterm or term delivery). We show that significant transcriptomic differences exist between amniotic fluid neutrophils and monocytes/macrophages from women with intra-amniotic infection, indicating the distinct roles these cells play. The transcriptome of amniotic fluid immune cells varies based on their maternal or fetal origin, and the significant transcriptomic differences between fetal and maternal monocytes/macrophages imply that those of fetal origin exhibit impaired functions. Notably, transcriptomic changes in amniotic fluid monocytes/macrophages suggest that these immune cells collaborate with neutrophils in the trafficking of fetal leukocytes throughout the umbilical cord (i.e., funisitis). Finally, amniotic fluid neutrophils and monocytes/macrophages from preterm deliveries display enhanced transcriptional activity compared to those from term deliveries, highlighting the protective role of these cells during this vulnerable period. Collectively, these findings demonstrate the underlying complexity of local innate immune responses in women with intra-amniotic infection and provide new insights into the functions of neutrophils and monocytes/macrophages in the amniotic cavity.


Assuntos
Âmnio/imunologia , Líquido Amniótico/imunologia , Corioamnionite/imunologia , Macrófagos/fisiologia , Neutrófilos/fisiologia , Trabalho de Parto Prematuro/imunologia , Gravidez/imunologia , Movimento Celular , Células Cultivadas , Feminino , Feto , Perfilação da Expressão Gênica , Humanos , Imunidade Inata , Análise de Sequência de RNA
7.
Int J Med Sci ; 17(16): 2496-2504, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33029092

RESUMO

Background: To maintain the normal pregnancy, suppression of inflammatory signaling pathway is a crucial physiologic response. Dexmedetomidine has been used for labor analgesia or supplement of inadequate regional analgesia during delivery. And it has been reported that dexmedetomidine has an anti-inflammatory effect. In this study, we examined the influence of dexmedetomidine on the expression of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2) and inflammatory cytokines in lipopolysaccharide (LPS)-stimulated human amnion-derived WISH cells. In addition, we evaluated the association of mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathway in anti-inflammatory effect of dexmedetomidine. Methods: Human amnion-derived WISH cells were pretreated with various concentrations of dexmedetomidine (0.001-1 µg/ml) for 1 h and after then treated with LPS (1 µg/ml) for 24 h. MTT assay was conducted to evaluate the cytotoxicity. Nitric oxide (NO) production was analyzed using Griess-reaction microassay. RT-PCR was performed for analysis of mRNA expressions of COX-2, PGE2, tumor necrosis factor (TNF)-α and interlukin (IL)-1ß. Protein expressions of COX-2, PGE2, p38 and NF-κB were analyzed by western blotting. Results: LPS and dexmedetomidine had no cytotoxic effect on WISH cells. There was no difference in NO production after dexmedetomidine pretreatment. The mRNA and protein expressions of COX-2 and PGE2 were decreased by dexmedetomidine pretreatment in LPS-treated WISH cells. Dexmedetomidine also attenuated the LPS-induced mRNA expression of TNF-α and IL-1ß. The activation of p38 and NF-κB was suppressed by dexmedetomidine pretreatment in LPS-treated WISH cells. Conclusion: We demonstrated that dexmedetomidine pretreatment suppressed the expressions of inflammatory mediators increased by LPS. In addition, this study suggests that anti-inflammatory effect of dexmedetomidine on WISH cells was mediated by the inhibitions of p38 and NF-κB activation.


Assuntos
Âmnio/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Dexmedetomidina/farmacologia , Inflamação/tratamento farmacológico , Âmnio/citologia , Âmnio/imunologia , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Ciclo-Oxigenase 2/metabolismo , Dexmedetomidina/uso terapêutico , Dinoprostona/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Front Immunol ; 11: 1899, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32983111

RESUMO

Background: Infection/inflammation is an important causal factor in spontaneous preterm birth (sPTB). Most mechanistic studies have concentrated on the role of bacteria, with limited focus on the role of viruses in sPTB. Murine studies support a potential multi-pathogen aetiology in which a double or sequential hit of both viral and bacterial pathogens leads to a higher risk preterm labour. This study aimed to determine the effect of viral priming on bacterial induced inflammation in human in vitro models of ascending and haematogenous infection. Methods: Vaginal epithelial cells, and primary amnion epithelial cells and myocytes were used to represent cell targets of ascending infection while interactions between peripheral blood mononuclear cells (PBMCs) and placental explants were used to model systemic infection. To model the effect of viral priming upon the subsequent response to bacterial stimuli, each cell type was stimulated first with a TLR3 viral agonist, and then with either a TLR2 or TLR2/6 agonist, and responses compared to those of each agonist alone. Immunoblotting was used to detect cellular NF-κB, AP-1, and IRF-3 activation. Cellular TLR3, TLR2, and TLR6 mRNA was quantified by RT-qPCR. Immunoassays were used to measure supernatant cytokine, chemokine and PGE2 concentrations. Results: TLR3 ("viral") priming prior to TLR2/6 agonist ("bacterial") exposure augmented the pro-inflammatory, pro-labour response in VECs, AECs, myocytes and PBMCs when compared to the effects of agonists alone. In contrast, enhanced anti-inflammatory cytokine production (IL-10) was observed in placental explants. Culturing placental explants in conditioned media derived from PBMCs primed with a TLR3 agonist enhanced TLR2/6 agonist stimulated production of IL-6 and IL-8, suggesting a differential response by the placenta to systemic inflammation compared to direct infection as a result of haematogenous spread. TLR3 agonism generally caused increased mRNA expression of TLR3 and TLR2 but not TLR6. Conclusion: This study provides human in vitro evidence that viral infection may increase the susceptibility of women to bacterial-induced sPTB. Improved understanding of interactions between viral and bacterial components of the maternal microbiome and host immune response may offer new therapeutic options, such as antivirals for the prevention of PTB.


Assuntos
Âmnio/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Miométrio/efeitos dos fármacos , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/virologia , Receptor 2 Toll-Like/agonistas , Receptor 3 Toll-Like/agonistas , Receptor 6 Toll-Like/agonistas , Vagina/efeitos dos fármacos , Âmnio/imunologia , Âmnio/metabolismo , Linhagem Celular , Citocinas/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Interações Hospedeiro-Patógeno , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Miométrio/imunologia , Miométrio/metabolismo , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/metabolismo , Transdução de Sinais , Técnicas de Cultura de Tecidos , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Receptor 6 Toll-Like/genética , Receptor 6 Toll-Like/metabolismo , Vagina/imunologia , Vagina/metabolismo
9.
Cells ; 9(9)2020 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-32961693

RESUMO

The immunoprivilege status characteristic of human amnion epithelial cells (hAECs) has been recently highlighted in the context of xenogenic transplantation. However, the mechanism(s) involved in such regulatory functions have been so far only partially been clarified. Here, we have analyzed the expression of HLA-Ib molecules in isolated hAEC obtained from full term placentae. Moreover, we asked whether these molecules are involved in the immunoregulatory functions of hAEC. Human amnion-derived cells expressed surface HLA-G and HLA-F at high levels, whereas the commonly expressed HLA-E molecule has been measured at a very low level or null on freshly isolated cells. HLA-Ib molecules can be expressed as membrane-bound and soluble forms, and in all hAEC batches analyzed we measured high levels of sHLA-G and sHLA-E when hAEC were maintained in culture, and such a release was time-dependent. Moreover, HLA-G was present in extracellular vesicles (EVs) released by hAEC. hAEC suppressed T cell proliferation in vitro at different hAEC:T cell ratios, as previously reported. Moreover, inhibition of T cell proliferation was partially reverted by pretreating hAEC with anti-HLA-G, anti-HLA-E and anti-ß2 microglobulin, thus suggesting that HLA-G and -E molecules are involved in hAEC-mediated suppression of T cell proliferation. Finally, either large-size EV (lsEV) or small-size EV (ssEV) derived from hAEC significantly modulated T-cell proliferation. In conclusion, we have here characterized one of the mechanism(s) underlying immunomodulatory functions of hAEC, related to the expression and release of HLA-Ib molecules.


Assuntos
Âmnio/imunologia , Comunicação Celular/imunologia , Células Epiteliais/imunologia , Antígenos HLA-G/genética , Antígenos de Histocompatibilidade Classe I/genética , Linfócitos T/imunologia , Âmnio/citologia , Anticorpos Monoclonais/farmacologia , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Células Epiteliais/citologia , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Regulação da Expressão Gênica , Antígenos HLA-G/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Cultura Primária de Células , Linfócitos T/citologia , Microglobulina beta-2/antagonistas & inibidores , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia , Antígenos HLA-E
10.
Front Immunol ; 11: 1645, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849565

RESUMO

Context and Objectives: Inflammation is the leading mechanism involved in both physiological and pathological rupture of fetal membranes. Our aim was to obtain a better characterization of the inflammasome-dependent inflammation processes in these tissues, with a particular focus on the nucleotide-binding oligomerization domain (NOD)-like receptor, pyrin domain containing protein 7 (NLRP7) inflammasome. Methods: The presence of NLRP7 inflammasome actors [NLRP7, apoptosis-associated speck-like protein containing a CARD domain (ASC), and caspase-1] was confirmed by reverse transcriptase-polymerase chain reaction (RT-PCR) in human amnion and choriodecidua at the three trimesters and at term. The protein concentrations were then determined by enzyme-linked immunosorbent assay in term tissues, with or without labor. The presence of Mycoplasma salivarium and Mycoplasma fermentans in human fetal membranes was investigated using a PCR approach. Human amnion epithelial cells (AECs) were treated for 4 or 20 h with fibroblast-stimulating lipopeptide-1 (FSL-1), a M. salivarium-derived ligand. Transcripts and proteins quantity was then measured by RT-quantitative PCR and Western blotting, respectively. NLRP7 and ASC colocalization was confirmed by immunofluorescence. Western blots allowed analysis of pro-caspase-1 and gasdermin D cleavage. Results: NLRP7, ASC, and caspase-1 transcripts were expressed in both sheets of human fetal membranes during all pregnancy stages, but only ASC protein expression was increased with labor. In addition, M. salivarium and M. fermentans were detected for the first time in human fetal membranes. NLRP7 and caspase-1 transcripts, as well as NLRP7, ASC, and pro-caspase-1 protein levels, were increased in FSL-1-treated AECs. The NLRP7 inflammasome assembled around the nucleus, and pro-caspase-1 and gasdermin D were cleaved into their mature forms after FSL-1 stimulation. Conclusion: Two new mycoplasmas, M. salivarium and M. fermentans, were identified in human fetal membranes, and a lipopeptide derived from M. salivarium was found to induce NLRP7 inflammasome formation in AECs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Âmnio/efeitos dos fármacos , Diglicerídeos/farmacologia , Células Epiteliais/efeitos dos fármacos , Inflamassomos/metabolismo , Mycoplasma fermentans/metabolismo , Mycoplasma salivarium/metabolismo , Oligopeptídeos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Âmnio/imunologia , Âmnio/metabolismo , Âmnio/microbiologia , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/genética , Caspase 1/metabolismo , Células Cultivadas , Cesárea , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos/genética , Mycoplasma fermentans/isolamento & purificação , Mycoplasma salivarium/isolamento & purificação , Parto , Gravidez , Trimestres da Gravidez , Transdução de Sinais
11.
Virology ; 548: 93-100, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32838950

RESUMO

Human cytomegalovirus (HCMV) infects the chorioamnion, but whether these infections cause fetal membrane dysfunction remains poorly understood. We sought to assess whether guinea pig cytomegalovirus (GPCMV) infects amnion-derived cells in vitro, compare the inflammatory response of amnion cells to GPCMV and HCMV, and determine if GPCMV infects the amnion in vivo. We found that GPCMV replicates in primary guinea pig amnion derived cells and HPV16 E6/E7-transduced amniotic epithelial cells (AEC[E6/E7]s). HCMV and GPCMV infection of amnion cells increased the transcription of the chemokines CCL5/Ccl5, CXCL8/Cxcl8, and CXCL10/Cxcl10. Myd88-knockdown decreased Ccl5 and Cxc8 transcription in GPCMV-infected AEC[E6/E7]s. GPCMV was detected in the guinea pig amnion after primary maternal infection, revealing that guinea pigs are an appropriate model to study fetal membrane physiology after cytomegalovirus infection. As inflammation is known to cause fetal membrane weakening, the amnion's response to cytomegalovirus infection may cause preterm birth and other adverse pregnancy outcomes.


Assuntos
Âmnio/imunologia , Quimiocinas/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Complicações na Gravidez/imunologia , Âmnio/virologia , Animais , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quimiocinas/genética , Citomegalovirus/genética , Citomegalovirus/imunologia , Infecções por Citomegalovirus/genética , Feminino , Cobaias , Humanos , Interleucina-8/genética , Interleucina-8/imunologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/virologia
12.
Front Immunol ; 11: 54, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117234

RESUMO

Human mesenchymal stromal/stem cells (MSCs), being immunoprivileged and having immunomodulatory ability, represent a promising tool to be applied in the field of regenerative medicine. Based on numerous in vitro evidences, the immunological effects of MSCs on immune cells could depend on different mechanisms as cell-to-cell contact and paracrine signals. Furthermore, recent studies have shown that the immunomodulatory activity of MSCs is initiated by activated immune cells; thus, their interaction represents a potential homeostatic mechanism by which MSCs regulate the immune response. MSCs also release exosomes able to give different effects, in a paracrine manner, by influencing inflammatory processes. In this study, we aimed to establish the potential role of human amnion-derived MSCs (hAMSCs), in immunomodulation. We found that the immunosuppressive properties of hAMSCs are not constitutive, but require "supportive signals" capable of promoting these properties. Indeed, we observed that hAMSCs alone are not able to produce an adequate amount of soluble immunomodulatory factors. Here, we studied, in depth, the strong immunomodulatory licensing signal deriving from the direct interaction between hAMSCs and stimulated peripheral blood mononuclear cells. We found that the immunomodulatory effect of hAMSCs also depends on cell-to-cell contact through the contribution of the PDL-1/PD-1 axis. We then investigated the IFN-γ priming of hAMSCs (γ-hAMSCs), which induce the increase of PDL-1 expression, high production of IDO, and upregulation of different immunomodulatory exosome-derived miRNAs. Our miRNA-target network analysis revealed that nine of the deregulated miRNAs are involved in the regulation of key proteins that control both T cell activation/anergy and monocyte differentiation pathways. Finally, we observed that γ-hAMSCs induce in monocytes both M2-like phenotype and the increase of IL-10 production. The extensive implications of MSCs in modulating different aspects of the immune system make these cells attractive candidates to be employed in therapeutic application in immune-based diseases. For these reasons, we aimed, with this study, to shed light on the potential of hAMSCs, and how they could become a useful tool for treating different inflammatory diseases, including end-stage pathologies or adverse effects in transplanted patients.


Assuntos
Âmnio/imunologia , Comunicação Celular/imunologia , Imunomodulação/imunologia , Ativação Linfocitária/imunologia , Células-Tronco Mesenquimais/imunologia , Âmnio/citologia , Antígeno B7-H1/metabolismo , Diferenciação Celular , Proliferação de Células , Técnicas de Cocultura , Citocinas/metabolismo , Exossomos/imunologia , Voluntários Saudáveis , Humanos , Quinase I-kappa B/metabolismo , Imunidade , Fatores Imunológicos/metabolismo , Terapia de Imunossupressão , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/farmacologia , Leucócitos Mononucleares/imunologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs , Monócitos , Cultura Primária de Células , Receptor de Morte Celular Programada 1/metabolismo
13.
J Gen Virol ; 101(4): 426-439, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32068527

RESUMO

Congenital cytomegalovirus (cCMV) is a leading cause of birth defects. The guinea pig is the only small cCMV animal model. Guinea pig cytomegalovirus (GPCMV) encodes similar glycoprotein complexes to human CMV (HCMV) including gB and the gH-based pentamer complex (PC). In HCMV, both gB and PC are neutralizing antibody antigens. The relevance of GPCMV PC for virus tropism and vaccine target remains controversial. A novel guinea pig placental amniotic sac epithelial (GPASE) cell-line did not express viral cell receptor platelet derived growth factor receptor alpha (PDGFRA) and resulted in requirement for the PC for GPCMV infection unless PDGFRA was ectopically expressed. High titer anti-gB sera from a GPCMV gB vaccine study was evaluated for GPCMV neutralizing capability on GPASE cells in comparison to convalescent sera from GPCMV(PC+) or GPCMV(PC-) infected animals. Anti-gB sera neutralized fibroblast infection but was less effective compared to anti-GPCMV(PC-), which had antibodies to gH/gL. However, both anti-GPCMV(PC-) and anti-gB sera similarly had reduced neutralizing capability on GPASE and renal epithelial cells in comparison to anti-GPCMV(PC+) sera, which had additional antibodies to PC. Overall, results demonstrate the importance of the PC for GPCMV tropism to various cell types that lack PDGFRA expression and the limited ability of anti-gB sera to neutralize GPCMV on non-fibroblast cells despite the essential nature of gB glycoprotein.


Assuntos
Âmnio/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/genética , Glicoproteínas/metabolismo , Placenta/imunologia , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/imunologia , Âmnio/citologia , Âmnio/metabolismo , Âmnio/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidade , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Feminino , Técnicas de Inativação de Genes , Cobaias , Mutação , Testes de Neutralização , Placenta/citologia , Placenta/metabolismo , Placenta/virologia , Gravidez , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Tropismo Viral
14.
J Immunol ; 203(7): 1793-1807, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31492740

RESUMO

Prematurity is the leading cause of perinatal morbidity and mortality worldwide. In most cases, preterm birth is preceded by spontaneous preterm labor, a syndrome that is associated with intra-amniotic inflammation, the most studied etiology. However, the remaining etiologies of preterm labor are poorly understood; therefore, most preterm births are categorized as idiopathic. In this study, we provide evidence showing that the fetal immune system undergoes premature activation in women with preterm labor without intra-amniotic inflammation, providing a potential new mechanism of disease for some cases of idiopathic preterm birth. First, we showed that fetal T cells are a predominant leukocyte population in amniotic fluid during preterm gestations. Interestingly, only fetal CD4+ T cells were increased in amniotic fluid of women who underwent idiopathic preterm labor and birth. This increase in fetal CD4+ T cells was accompanied by elevated amniotic fluid concentrations of T cell cytokines such as IL-2, IL-4, and IL-13, which are produced by these cells upon in vitro stimulation, but was not associated with the prototypical cytokine profile observed in women with intra-amniotic inflammation. Also, we found that cord blood T cells, mainly CD4+ T cells, obtained from women with idiopathic preterm labor and birth displayed enhanced ex vivo activation, which is similar to that observed in women with intra-amniotic inflammation. Finally, we showed that the intra-amniotic administration of activated neonatal CD4+ T cells induces preterm birth in mice. Collectively, these findings provide evidence suggesting that fetal T cell activation is implicated in the pathogenesis of idiopathic preterm labor and birth.


Assuntos
Âmnio/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/imunologia , Feto/imunologia , Ativação Linfocitária , Trabalho de Parto Prematuro/imunologia , Adulto , Âmnio/patologia , Animais , Linfócitos T CD4-Positivos/patologia , Feminino , Feto/patologia , Humanos , Camundongos , Trabalho de Parto Prematuro/patologia , Gravidez
15.
Reproduction ; 158(1): 95-107, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31022702

RESUMO

Inflammation plays a pivotal role in the terminal process of human labor and delivery, including myometrial contractions and membrane rupture. TNF-alpha-induced protein 8-like-2 (TIPE2) is a novel inflammation regulator; however, there are no studies on the role of TIPE2 in human labor. We report that in myometrium, there is decreased TIPE2 mRNA expression during late gestation which was further decreased in labor. In fetal membranes, TIPE2 mRNA expression was decreased with both term and preterm labor compared to no labor samples. Knockdown of TIPE2 by siRNA in primary myometrium and amnion cells was associated with an augmentation of IL1B and TNF-induced expression of pro-inflammatory cytokines and chemokines; expression of contraction-associated proteins and secretion of the uterotonic prostaglandin PGF2α and expression of extracellular matrix degrading enzymes. In TIPE2-deficient myometrial cells treated with inhibitors of NF-κB or ERK1/2, the secretion of pro-labor mediators was reduced back to control levels. In conclusion, these in vitro experiments indicate that loss of TIPE2 exacerbates the inflammatory response.


Assuntos
Âmnio/efeitos dos fármacos , Anti-Inflamatórios/administração & dosagem , Inflamação/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , Trabalho de Parto/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miométrio/efeitos dos fármacos , Adulto , Âmnio/imunologia , Âmnio/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Trabalho de Parto/imunologia , Trabalho de Parto/metabolismo , Miométrio/imunologia , Miométrio/metabolismo , NF-kappa B/metabolismo , Gravidez
16.
Genet Test Mol Biomarkers ; 22(12): 665-673, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30481073

RESUMO

BACKGROUND: Human amniotic membrane-derived mesenchymal stem cells (hAM-MSCs) are considered a new and favorable source of stem cells for cell replacement-based therapy. Some microRNAs (miRNAs) have been reported to participate in the regulation of immune responses. Our aim was to investigate the effects of miR-21 on the biological characteristics, immunoregulatory properties, and potential mechanisms of hAM-MSCs. METHODS: hAM-MSCs were isolated from the placental amnion membrane of a newborn. Cell proliferation, cell cycle, apoptosis, and expressions of cell surface markers were measured by CCK-8 and flow cytometric assays in hAM-MSCs. The expression of mesenchymal-specific antigens vimentin and stage-specific embryonic antigen-4 (SSEA-4) were identified by immunofluorescence staining. Tumor necrosis factor alpha (TNF-α), monocyte chemotactic protein-1 (MCP-1), and interleukin-10 (IL-10) expressions in the cocultured supernatant of hAM-MSCs and peripheral blood mononuclear cells (PBMC) were detected via enzyme-linked immunosorbent assays (ELISA). RESULTS: Flow cytometric analyses revealed that the positive expression rates of the cell surface markers CD29, CD44, CD73, and CD90 in hAM-MSCs were 97.3%, 96.3%, 97.8%, and 98.2%, respectively, while the rates of CD34 and CD45 expression were only 0.6% and 0.84%, respectively. The immunofluorescent staining results showed that vimentin and SSEA-4 were positive in hAM-MSCs. CCK-8 assays revealed that miR-21 overexpression significantly promoted hAM-MSC proliferation. Cell cycle analyses revealed that the number of hAM-MSCs-miR-21+ cells during the synthesis phase (S phase) was significantly increased. miR-21 overexpression also significantly inhibited apoptosis in hAM-MSCs. The ELISA analyses revealed that miR-21 overexpression enhanced the inhibitory effect of hAM-MSCs on the secretion of TNF-α and MCP-1 as well as the promotive effect on the secretion of IL-10 in PBMC cocultured with miR-21-hAM-MSCs. In addition, miR-21 downregulation reduced the inhibitory effect of hAM-MSCs on the secretion of TNF-α, MCP-1, and the promotive effect on the secretion of IL-10 in PBMC cocultured with anti-miR-21-hAM-MSCs. CONCLUSIONS: Our data showed that miR-21 promoted hAM-MSCs proliferation, inhibited apoptosis, and was involved in controlling the immunoregulatory capacity of hAM-MSCs.


Assuntos
Âmnio/imunologia , Células-Tronco Mesenquimais/imunologia , MicroRNAs/imunologia , Antígenos de Superfície/metabolismo , Apoptose , Ciclo Celular , Proliferação de Células , Células Cultivadas , Quimiocina CCL2/biossíntese , Regulação para Baixo , Citometria de Fluxo , Humanos , Recém-Nascido , Interleucina-10/biossíntese , MicroRNAs/genética , Sincalida/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima
17.
Am J Reprod Immunol ; 80(5): e13049, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30225853

RESUMO

PROBLEM: The inflammasome is implicated in the mechanisms that lead to spontaneous preterm labor (PTL). However, whether there is inflammasome activation in the amniotic cavity of women with PTL and intra-amniotic infection (IAI) or sterile intra-amniotic inflammation (SIAI) is unknown. METHOD OF STUDY: Amniotic fluid samples were collected from women with PTL who delivered at term (n = 31) or preterm without IAI or SIAI (n = 35), with SIAI (n = 27), or with IAI (n = 17). As a readout of inflammasome activation, extracellular ASC (apoptosis-associated speck-like protein containing a CARD) was measured in amniotic fluid by ELISA and the expression of ASC, caspase-1, and interleukin (IL)-1ß was detected in the chorioamniotic membranes by multiplex immunofluorescence. Acute inflammatory responses in amniotic fluid and the placenta were also evaluated. RESULTS: (a) Amniotic fluid concentrations of ASC and IL-6 were higher in women with PTL and IAI or SIAI than in those who delivered preterm or at term without intra-amniotic inflammation; (b) amniotic fluid concentrations of ASC and IL-6 were lower in women with PTL and SIAI than in those with IAI; (c) there was a significant nonlinear correlation between ASC and IL-6 amniotic fluid concentrations; (d) the expression of inflammasome-related proteins (ASC, caspase-1, and IL-1ß) in the chorioamniotic membranes was increased in women with PTL and IAI or SIAI than in those who delivered preterm or at term without intra-amniotic inflammation; (e) inflammasome activation in the chorioamniotic membranes was weaker in women with PTL and SIAI than in those with IAI; (f) women with PTL and IAI had elevated amniotic fluid white blood cell counts compared to those without this clinical condition; and (g) severe acute placental inflammatory lesions were observed in women with PTL and IAI and in a subset of women with PTL and SIAI. CONCLUSION: Inflammasome activation occurs in the settings of intra-amniotic infection and sterile intra-amniotic inflammation during spontaneous preterm labor.


Assuntos
Aborto Espontâneo/imunologia , Âmnio/imunologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Infecções/imunologia , Inflamassomos/metabolismo , Inflamação/imunologia , Placenta/imunologia , Adulto , Líquido Amniótico/metabolismo , Caspase 1/metabolismo , Estudos Transversais , Feminino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Gravidez , Estudos Retrospectivos , Adulto Jovem
18.
J Reprod Immunol ; 127: 24-35, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29751216

RESUMO

Preterm birth is the primary cause of neonatal deaths and morbidities. Pathological processes causally linked to preterm birth are inflammation and infection. Pellino-1 (Peli1) has previously been found to regulate the inflammatory response in non-gestational tissues in response to toll-like receptor (TLR) ligands and pro-inflammatory cytokines. The aims of this study were to determine the effect of labor on Peli1 expression in myometrium and fetal membranes, and the effect of Peli1 silencing by siRNA (siPELI1) on the production of pro-inflammatory and pro-labor mediators. The expression of Peli1 was found to be higher in myometrium and fetal membranes with term labor, compared to non-laboring samples. Peli1 mRNA and protein expression was also higher in amnion from women with preterm histological chorioamnionitis. In human primary myometrial cells, siPELI1 transfected cells showed a decrease in pro-inflammatory cytokine IL6, chemokines (CXCL8, CCL2) and adhesion molecule ICAM1 when in the presence of pro-inflammatory cytokine TNF, TLR2/6 ligand fsl-1, TLR5 ligand flagellin, and TLR3 ligand poly(I:C). Similarly in primary amnion cells, siPELI1 transfected cells decreased IL1B-induced expression and secretion of IL6 and CXCL8. In siPELI1 transfected myometrial cells, there was a decrease in prostaglandin PGF2α and its receptor, PTGFR mRNA expression when treated with TNF. There was a decrease in NF-κB RELA transcriptional activity in siPELI1 transfected cells in the presence of TNF, fsl-1 and flagellin, but not poly(I:C). Our study suggests a novel role for Peli1 in regulating pro-inflammatory and pro-labor mediators through TNF and TLR signalling.


Assuntos
Âmnio/imunologia , Membranas Extraembrionárias/fisiologia , Inflamação/imunologia , Trabalho de Parto/fisiologia , Miométrio/fisiologia , Proteínas Nucleares/metabolismo , Nascimento Prematuro/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Âmnio/citologia , Feminino , Humanos , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Proteínas Nucleares/genética , Gravidez , Nascimento Prematuro/genética , Nascimento Prematuro/imunologia , RNA Interferente Pequeno/genética , Transdução de Sinais , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética
19.
Reprod Biol ; 18(2): 182-188, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29729842

RESUMO

Unexplained recurrent spontaneous abortion (URSA) has been assumed to be caused by a defect in maternal immunological tolerance to the fetus. Human amniotic epithelial cells (hAECs) have stem cell-like features and the ability to modulate the innate and adoptive immune responses. This study aimed to investigate whether hAECs have immunomodulatory effects on naive CD4+ T cells from URSA patients. hAECs were obtained from 15 healthy pregnant women and phenotypic profile of hAECs was determined by flow cytometry. Naive CD4+ T cells were isolated from 25 URSA patients using an immunomagnetic separation method. Naive T cells were stimulated with anti-CD3/anti-CD28 antibodies and co-cultured with different numbers of hAECs for 3 and 6 days. Immunomodulatory effect of hAECs on activation of stimulated T cell was assessed by flow cytometry and Enzyme-linked immunoasorbent assay (ELISA). The hAECs effect on pro-inflammatory cytokines production of activated T cells was also measured by ELISA. Our results indicated that hAECs significantly inhibited the activation of naive T cells in a dose-dependent manner (p < 0.0001-0.05). They significantly reduced the production of transforming growth factor-beta1 (TGF-ß1) of stimulated CD4+T cells (p < 0.0001-0.05). Moreover, hAECs had potent immunomodulatory effects on the production of interferon-gamma (IFN-γ) and interleukin-17A (IL-17A) of activated T cells (p < 0.0001-0.01). These findings suggest that hAECs may be a suitable cell source to modulate abnormal immune responses in women with URSA.


Assuntos
Aborto Habitual/imunologia , Âmnio/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Inflamação/imunologia , Aborto Habitual/metabolismo , Adulto , Âmnio/citologia , Âmnio/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Gravidez , Adulto Jovem
20.
Cell Transplant ; 27(1): 45-54, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29562776

RESUMO

Cell-derived microvesicles (MVs) are a recently discovered mechanism of cell-to-cell communication. Our previous data show that MVs secreted by equine amniotic mesenchymal-derived cells (AMCs) are involved in downregulation of proinflammatory genes in lipopolysaccharide-stressed equine tendon and endometrial cells. The aim of the present study was to evaluate whether AMC-MVs contain selected microRNAs (miRNAs) involved in inflammation. Two pools of cells, derived from 3 amniotic membranes each, and their respective MVs were collected. Small RNAs were extracted and deep sequenced, followed by miRNA in silico detection. The analysis identified 1,285 miRNAs, which were quantified both in AMCs and MVs. Among these miRNAs, 401 were classified as Equus caballus miRNAs, 257 were predicted by homology with other species (cow, sheep, and goat), and 627 were novel candidate miRNAs. Moreover, 146 miRNAs differentially expressed (DE) in AMCs and MVs were identified, 36 of which were known and the remaining were novel. Among the known DE miRNAs, 17 showed higher expression in MVs. Three of these were validated by real time polymerase chain reaction: eca-miR-26, eca-miR-146a, and eca-miR-223. Gene ontology analysis of validated targets showed that the DE miRNAs in cells and MVs could be involved both in immune system regulation by modulating interleukin signaling and in the inflammatory process. In conclusion, this study suggests a significant role of AMCs in modulating immune response through cell-cell communication via MV-shuttling miRNAs.


Assuntos
Âmnio/metabolismo , MicroRNAs/metabolismo , Âmnio/imunologia , Animais , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Cavalos , Lipopolissacarídeos/farmacologia , MicroRNAs/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...