Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38791199

RESUMO

Danger-associated molecular patterns (DAMPs) are elevated within the amniotic cavity, and their increases correlate with advancing gestational age, chorioamnionitis, and labor. Although the specific triggers for their release in utero remain unclear, it is thought that they may contribute to the initiation of parturition by influencing cellular stress mechanisms that make the fetal membranes (FMs) more susceptible to rupture. DAMPs induce inflammation in many different tissue types. Indeed, they precipitate the subsequent release of several proinflammatory cytokines that are known to be key for the weakening of FMs. Previously, we have shown that in vitro stretch of human amnion epithelial cells (hAECs) induces a cellular stress response that increases high-mobility group box-1 (HMGB1) secretion. We have also shown that cell-free fetal DNA (cffDNA) induces a cytokine response in FM explants that is fetal sex-specific. Therefore, the aim of this work was to further investigate the link between stretch and the DAMPs HMGB1 and cffDNA in the FM. These data show that stretch increases the level of cffDNA released from hAECs. It also confirms the importance of the sex of the fetus by demonstrating that female cffDNA induced more cellular stress than male fetuses. Our data treating hAECs and human amnion mesenchymal cells with HMGB1 show that it has a differential effect on the ability of the cells of the amnion to upregulate the proinflammatory cytokines and propagate a proinflammatory signal through the FM that may weaken it. Finally, our data show that sulforaphane (SFN), a potent activator of Nrf2, is able to mitigate the proinflammatory effects of stretch by decreasing the levels of HMGB1 release and ROS generation after stretch and modulating the increase of key cytokines after cell stress. HMGB1 and cffDNA are two of the few DAMPs that are known to induce cytokine release and matrix metalloproteinase (MMP) activation in the FMs; thus, these data support the general thesis that they can function as potential central players in the normal mechanisms of FM weakening during the normal distension of this tissue at the end of a normal pregnancy.


Assuntos
Membranas Extraembrionárias , Proteína HMGB1 , Inflamação , Humanos , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Feminino , Gravidez , Inflamação/metabolismo , Inflamação/patologia , Membranas Extraembrionárias/metabolismo , Ácidos Nucleicos Livres/metabolismo , Masculino , Âmnio/metabolismo , Citocinas/metabolismo , Células Epiteliais/metabolismo , Células Cultivadas , Alarminas/metabolismo
2.
Int J Mol Sci ; 23(12)2022 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-35742804

RESUMO

The receptor of advanced glycation end products (RAGE) is a receptor that is thought to be a key driver of inflammation in pregnancy, SARS-CoV-2, and also in the comorbidities that are known to aggravate these afflictions. In addition to this, vulnerable populations are particularly susceptible to the negative health outcomes when these afflictions are experienced in concert. RAGE binds a number of ligands produced by tissue damage and cellular stress, and its activation triggers the proinflammatory transcription factor Nuclear Factor Kappa B (NF-κB), with the subsequent generation of key proinflammatory cytokines. While this is important for fetal membrane weakening, RAGE is also activated at the end of pregnancy in the uterus, placenta, and cervix. The comorbidities of hypertension, cardiovascular disease, diabetes, and obesity are known to lead to poor pregnancy outcomes, and particularly in populations such as Native Hawaiians and Pacific Islanders. They have also been linked to RAGE activation when individuals are infected with SARS-CoV-2. Therefore, we propose that increasing our understanding of this receptor system will help us to understand how these various afflictions converge, how forms of RAGE could be used as a biomarker, and if its manipulation could be used to develop future therapeutic targets to help those at risk.


Assuntos
COVID-19 , Produtos Finais de Glicação Avançada , Proteínas de Transporte , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Humanos , NF-kappa B/metabolismo , Gravidez , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptores Imunológicos/metabolismo , SARS-CoV-2
3.
Biomolecules ; 12(6)2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35740891

RESUMO

Nuclear-factor-E2-related factor 2 (Nrf2) is a key transcription factor for the regulation of cellular responses to cellular stress and inflammation, and its expression is significantly lower after spontaneous term labor in human fetal membranes. Pathological induction of inflammation can lead to adverse pregnancy outcomes such as pre-eclampsia, preterm labor, and fetal death. As stretch forces are known to act upon the fetal membranes in utero, we aimed to ascertain the effect of stretch on Nrf2 to increase our understanding of the role of this stimulus on cells of the amnion at term. Our results indicated a significant reduction in Nrf2 expression in stretched isolated human amnion epithelial cells (hAECs) that could be rescued with sulforaphane treatment. Downregulation of Nrf2 as a result of stretch was accompanied with activation of proinflammatory nuclear factor-kB (NF-kB) and increases in LDH activity, ROS, and HMGB1. This work supports stretch as a key modulator of cellular stress and inflammation in the fetal membranes. Our results showed that the modulation of the antioxidant response pathway in the fetal membranes through Nrf2 activation may be a viable approach to improve outcomes in pregnancy.


Assuntos
Âmnio , Fator 2 Relacionado a NF-E2 , Estresse Mecânico , Regulação para Baixo , Feminino , Humanos , Recém-Nascido , Inflamação/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Gravidez
4.
Am J Reprod Immunol ; 85(1): e13328, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32851715

RESUMO

A proinflammatory response driven by high-mobility group box 1 (HMGB1) is important for the success of both the early stages of pregnancy and parturition initiation. However, the tight regulation of HMGB1 within these two stages is critical, as increased HMGB1 can manifest into pregnancy-related pathologies. Although during the early stages of pregnancy HMGB1 is critical for the development and implantation of the embryo, and uterine decidualization, high levels within the uterine cavity have been linked to pregnancy failure. In addition, chronic inflammation, resultant from increased HMGB1 within the maternal circulation and gestational tissues, also increases the risk for preterm labor, preterm birth, or infant mortality. Due to the link between HMGB1 and several pregnancy pathologies, the possibility of leveraging HMGB1 as a biomarker has been assessed. However, data are limited that demonstrate how known HMGB1 inhibitors could reduce inflammation within pregnancy. Thus, further research is warranted to improve our understanding of the potential of HMGB1 as a therapeutic target to reduce inflammation within pregnancy. This review aims to describe what is understood about the role of HMGB1 that drives inflammation throughout pregnancy and highlight its potential as a biomarker and therapeutic target within this context.


Assuntos
Proteína HMGB1/imunologia , Inflamação/imunologia , Complicações na Gravidez/imunologia , Animais , Biomarcadores , Feminino , Proteína HMGB1/química , Humanos , Gravidez
5.
Front Physiol ; 11: 602, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32625109

RESUMO

The idea that cellular stress (including that precipitated by stretch), plays a significant role in the mechanisms initiating parturition, has gained considerable traction over the last decade. One key consequence of this cellular stress is the increased production of Danger Associated Molecular Patterns (DAMPs). This diverse family of molecules are known to initiate inflammation through their interaction with Pattern Recognition Receptors (PRRs) including, Toll-like receptors (TLRs). TLRs are the key innate immune system surveillance receptors that detect Pathogen Associated Molecular Patterns (PAMPs) during bacterial and viral infection. This is also seen during Chorioamnionitis. The activation of TLR commonly results in the activation of the pro-inflammatory transcription factor Nuclear Factor Kappa-B (NF-kB) and the downstream production of pro-inflammatory cytokines. It is thought that in the human fetal membranes both DAMPs and PAMPs are able, perhaps via their interaction with PRRs and the induction of their downstream inflammatory cascades, to lead to both tissue remodeling and weakening. Due to the high incidence of infection-driven Pre-Term Birth (PTB), including those that have preterm Premature Rupture of the Membranes (pPROM), the role of TLR in fetal membranes with Chorioamnionitis has been the subject of considerable study. Most of the work in this field has focused on the effect of PAMPs on whole pieces of fetal membrane and the resultant inflammatory cascade. This is important to understand, in order to develop novel prevention, detection, and therapeutic approaches, which aim to reduce the high number of mothers suffering from infection driven PTB, including those with pPROM. Studying the role of sterile inflammation driven by these endogenous ligands (DAMPs) activating PRRs system in the mesenchymal and epithelial cells in the amnion is important. These cells are key for the maintenance of the integrity and strength of the human fetal membranes. This review aims to (1) summarize the knowledge to date pertinent to the role of DAMPs and PRRs in fetal membrane weakening and (2) discuss the clinical potential brought by a better understanding of these pathways by pathway manipulation strategies.

6.
Placenta ; 44: 69-79, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27452440

RESUMO

INTRODUCTION: Infection accounts for over 40% of preterm premature rupture of the fetal membranes (PPROM), a major cause of preterm birth. Toll-like receptors (TLR) play key roles in pathogen surveillance but their expression and function in amnion mesenchymal cells (AMC) is unclear. The aims of this study were to determine the expression of all TLR isoforms and the effect of macrophage-activating lipoprotein-2 (MALP-2), derived from a common pathogen involved in PPROM, on human AMC. METHODS: AMC were isolated from normal, term amnion from repeat cesarean section. Semi-quantitative RT-PCR, immunocytochemistry, immunohistochemistry and western blotting were used to detect TLR isoform expression. Immunocytochemistry of NF-κB p65, pro-inflammatory cytokine secretion (ELISA), MTT assay, LDH assay, immunoblotting of cytosolic cytochrome c and cleaved caspase-3, and expression of 84 microRNAs by Qiagen miRNA PCR array were used to determine the functional effect of MALP-2 on AMC. RESULTS: TLR1-10 was detected in AMC, and protein expression of TLR2, 4, and 6 were confirmed. MALP-2 induced nuclear translocation of p65, reaching significance after 45 min (ANOVA, P < 0.05). MALP-2 did not cause apoptosis but did lead to significant secretion of IL-4, IL-6, and IL-8 (P < 0.05, 0.01, 0.001, respectively) and significant changes in miRNA-320a and miRNA-18a (P < 0.05). DISCUSSION: These results suggest that AMC elicit a pro-inflammatory response following stimulation with the known TLR2/6 ligand MALP-2. This data supports the idea that AMC express the innate immune system receptors that could help with immune surveillance during infection and contribute to inflammatory responses that lead to PPROM.


Assuntos
Âmnio/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Lipopeptídeos/farmacologia , Receptores Toll-Like/metabolismo , Âmnio/citologia , Âmnio/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 3/metabolismo , Feminino , Humanos , Gravidez , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/genética , Fator de Transcrição RelA/metabolismo
7.
Toxicol In Vitro ; 29(1): 103-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25283089

RESUMO

Human primary placental explant culture is well established for cytokine signaling and toxicity, but has not been validated for steroidogenic or metabolic toxicology. The technique has never been investigated in the mouse. We characterized human and mouse placental explants for up to 96 h in culture. Explant viability (Lactate dehydrogenase) and sex steroid levels were measured in media using spectrophotometry and ELISA, respectively. Expression and activities of the steroidogenic (3ß-hydroxysteroid dehydrogenase, Cytochrome P45017A1, Cytochrome P45019), conjugation (UDP-glucuronosyltransferase, sulfotransferase (SULT)), and regeneration (ß-glucuronidase, arylsulfatase C (ASC)) enzymes were determined biochemically in tissues with fluorimetric and spectrophotometric assays, and western blot. Explants were viable up to 96 h, but progesterone, estrone, and 17ß-estradiol secretion decreased. Steroidogenic enzyme expression and activities were stable in mouse explants and similar to levels in freshly isolated tissues, but were lower in human explants than in fresh tissue (P<0.01). Human and mouse explants exhibited significantly less conjugation after 96 h, SULT was not detected in the mouse, and neither explants had active ASC, although proteins were expressed. Mouse explants may be useful for steroid biochemistry and endocrine disruption studies, but not metabolic conjugation. In contrast, human explants may be useful for studying conjugation for <48 h, but not for steroid/endocrine studies.


Assuntos
Hormônios Esteroides Gonadais/antagonistas & inibidores , Placenta/efeitos dos fármacos , Testes de Toxicidade/métodos , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Aromatase/metabolismo , Estradiol/análise , Estradiol/biossíntese , Antagonistas de Estrogênios/efeitos adversos , Estrona/análise , Estrona/antagonistas & inibidores , Estrona/biossíntese , Feminino , Glucuronosiltransferase/metabolismo , Hormônios Esteroides Gonadais/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Placenta/química , Placenta/metabolismo , Gravidez , Progesterona/análise , Progesterona/antagonistas & inibidores , Progesterona/biossíntese , Esteroide 17-alfa-Hidroxilase/metabolismo , Técnicas de Cultura de Tecidos/métodos
8.
Eur J Drug Metab Pharmacokinet ; 40(4): 471-80, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25465229

RESUMO

Placental UDP-glucuronosyltransferase (UGT) enzymes have critical roles in hormone, nutrient, chemical balance and fetal exposure during pregnancy. Placental UGT1A isoforms were profiled and differences between preeclamptic (PE) and non-PE placental UGT expression determined. In third trimester villous placenta, UGT1A1, 1A4, 1A6 and 1A9 were expressed and active in all specimens (n = 10), but UGT1A3, 1A5, 1A7, 1A8 and 1A10 were absent. The UGT1A activities were comparable to human liver microsomes per milligram, but placental microsome yields were only 2 % of liver (1 mg/g of tissue vs. 45 mg/g of tissue). For successful PCR, placental collection and processing within 60 min from delivery, including DNAse and ≥300 ng of RNA in reverse transcription were essential and snap freezing in liquid nitrogen immediately was the best preservation method. Although UGT1A6 mRNA was lower in PE (P < 0.001), there were no other significant effects on UGT mRNA, protein or activities. A more comprehensive tissue sample set is required for confirmation of PE interactions with UGT. Placental UGT1A enzyme expression patterns are similar to the liver and a detoxicative role for placental UGT1A is inferred.


Assuntos
Glucuronosiltransferase/biossíntese , Placenta/enzimologia , Pré-Eclâmpsia/enzimologia , Nascimento a Termo/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Humanos , Placenta/patologia , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/genética , Gravidez
9.
Reprod Sci ; 14(8 Suppl): 35-41, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18089608

RESUMO

In the third trimester of normal pregnancy, the human fetal membranes become increasingly distended and use mechanotransduction and its downstream signaling to remodel and function. Their overdistension either by multifetal pregnancy or by polyhydramnios often leads to preterm birth, but the mechanism is unclear. Stretching of the fetal membranes in vitro upregulates several cytokines and enzymes that can drive collagen degradation, leading to membrane rupture. The sensitivity of this response appears to be specific for different cell types and is likely to result from differential activation of some key transcription factors and cofactors. Few cytokines in the fetal membranes respond to stretch: the most robust of these is pre-B-cell colony-enhancing factor (PBEF). This is constitutively expressed and protects the amnion cells from apoptosis caused by chronic static distension. However, it can also be stimulated by inflammation, infection, and hypoxia and upregulates a number of proinflammatory cytokines, chemokines, and enzymes important in the initiation of parturition. Therefore, it is proposed here that PBEF functions in normal pregnancy to protect the amnion cells as they become increasingly stretched, but if stimulated, it can initiate key events leading to parturition.


Assuntos
Citocinas/metabolismo , Membranas Extraembrionárias/metabolismo , Mecanorreceptores/metabolismo , Mecanotransdução Celular , Parto , Colágeno/metabolismo , Citocinas/imunologia , Membranas Extraembrionárias/enzimologia , Feminino , Ruptura Prematura de Membranas Fetais/metabolismo , Ruptura Prematura de Membranas Fetais/fisiopatologia , Humanos , Imunidade Inata , Mecanorreceptores/embriologia , Nicotinamida Fosforribosiltransferase/imunologia , Nicotinamida Fosforribosiltransferase/metabolismo , Poli-Hidrâmnios/metabolismo , Poli-Hidrâmnios/fisiopatologia , Gravidez , Terceiro Trimestre da Gravidez , Ruptura , Resistência à Tração , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa