Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunol Rev ; 308(1): 149-167, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35285967

RESUMO

Human parturition at term and preterm is an inflammatory process synchronously executed by both fetomaternal tissues to transition them from a quiescent state t an active state of labor to ensure delivery. The initiators of the inflammatory signaling mechanism can be both maternal and fetal. The placental (fetal)-maternal immune and endocrine mediated homeostatic imbalances and inflammation are well reported. However, the fetal inflammatory response (FIR) theories initiated by the fetal membranes (amniochorion) at the choriodecidual interface are not well established. Although immune cell migration, activation, and production of proparturition cytokines to the fetal membranes are reported, cellular level events that can generate a unique set of inflammation are not well discussed. This review discusses derangements to fetal membrane cells (physiologically and pathologically at term and preterm, respectively) in response to both endogenous and exogenous factors to generate inflammatory signals. In addition, the mechanisms of inflammatory signal propagation (fetal signaling of parturition) and how these signals cause immune imbalances at the choriodecidual interface are discussed. In addition to maternal inflammation, this review projects FIR as an additional mediator of inflammatory overload required to promote parturition.


Assuntos
Trabalho de Parto , Placenta , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Recém-Nascido , Inflamação/metabolismo , Trabalho de Parto/metabolismo , Parto/metabolismo , Placenta/metabolismo , Gravidez
2.
Int J Mol Sci ; 21(20)2020 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-33092043

RESUMO

The fetal inflammatory response, a key contributor of infection-associated preterm birth (PTB), is mediated by nuclear factor kappa B (NF-kB) activation. Na+/H+ exchanger regulatory factor-1 (NHERF1) is an adapter protein that can regulate intracellular signal transduction and thus influence NF-kB activation. Accordingly, NHERF1 has been reported to enhance proinflammatory cytokine release and amplify inflammation in a NF-kB-dependent fashion in different cell types. The objective of this study was to examine the role of NHERF1 in regulating fetal membrane inflammation during PTB. We evaluated the levels of NHERF1 in human fetal membranes from term labor (TL), term not in labor (TNIL), and PTB and in a CD1 mouse model of PTB induced by lipopolysaccharide (LPS). Additionally, primary cultures of fetal membrane cells were treated with LPS, and NHERF1 expression and cytokine production were evaluated. Gene silencing methods using small interfering RNA targeting NHERF1 were used to determine the functional relevance of NHERF1 in primary cultures. NHERF1 expression was significantly (p < 0.001) higher in TL and PTB membranes compared to TNIL membranes, and this coincided with enhanced (p < 0.01) interleukin (IL)-6 and IL-8 expression levels. LPS-treated animals delivering PTB had increased levels of NHERF1, IL-6, and IL-8 compared to phosphate-buffered saline (PBS; control) animals. Silencing of NHERF1 expression resulted in a significant reduction in NF-kB activation and IL-6 and IL-8 production as well as increased IL-10 production. In conclusion, downregulation of NHERF1 increased anti-inflammatory IL-10, and reducing NHERF1 expression could be a potential therapeutic strategy to reduce the risk of infection/inflammation associated with PTB.


Assuntos
Membranas Extraembrionárias/metabolismo , Inflamação/metabolismo , Fosfoproteínas/metabolismo , Nascimento Prematuro/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Membranas Extraembrionárias/patologia , Feminino , Humanos , Recém-Nascido , Lipopolissacarídeos , NF-kappa B/metabolismo , Fosfoproteínas/genética , Gravidez , Nascimento Prematuro/induzido quimicamente , RNA Interferente Pequeno , Trocadores de Sódio-Hidrogênio/genética
3.
Cytokine ; 81: 82-7, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26954343

RESUMO

BACKGROUND: High Mobility Group Box-1 (HMGB1) is considered a prototype alarmin molecule. Upon its extracellular release, HMGB1 engages pattern recognition receptors and the Receptor for Advanced Glycation End-products (RAGE) followed by an outpouring of inflammatory cytokines, including interleukin (IL)-6. METHODS: We assayed the amniotic fluid (AF) levels of HMGB1 and IL-6 in 255 women that either had a normal pregnancy outcome or delivered preterm. Immunohistochemistry on fetal membranes was used for cellular localization and validation of immunoassay findings. HMGB1 also was analyzed in amniochorion tissue explants subjected to endotoxin. RESULTS: AF HMGB1 levels are not gestational age regulated but are increased in women with intra-amniotic inflammation and preterm birth. The likely source is the damaged amniochorion, as demonstrated by immunohistochemistry and explant experiments. CONCLUSIONS: Our research supports a role for HMGB1 in the inflammatory response leading to preterm birth. As a delayed phase cytokine, in utero exposure to elevated AF HMGB1 levels may have an impact on the newborn beyond the time of birth.


Assuntos
Líquido Amniótico/metabolismo , Corioamnionite/metabolismo , Membranas Extraembrionárias/metabolismo , Proteína HMGB1/análise , Nascimento Prematuro/metabolismo , Adulto , Corioamnionite/fisiopatologia , Membranas Extraembrionárias/efeitos dos fármacos , Membranas Extraembrionárias/patologia , Feminino , Idade Gestacional , Proteína HMGB1/metabolismo , Humanos , Imunoensaio , Imuno-Histoquímica , Recém-Nascido , Interleucina-6/análise , Lipopolissacarídeos/farmacologia , Gravidez , Nascimento Prematuro/fisiopatologia , Receptor para Produtos Finais de Glicação Avançada/análise , Adulto Jovem
4.
Am J Obstet Gynecol ; 211(1): 48.e1-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24495670

RESUMO

OBJECTIVE: To test the effect of prolactin (PRL) on expression of proinflammatory cytokines and matrix metallopeptidase 9 (MMP-9) in vitro. STUDY DESIGN: Tissue explants were incubated from 4 to 48 hours alone or in the presence of 500 ng/mL PRL, and mRNA expression in tissues and secretion of interleukin (IL)-1ß, tumor necrosis factor alpha (TNF-α), MMP-2, and MMP-9 was quantified. RESULTS: Fetal membranes secreted IL-1ß, TNF-α, and MMP-9 in culture with consistent low concentration during the first 24 hours and then increased progressively. The presence of PRL during explant incubation significantly decreased the patterns of IL-1ß, TNF-α and MMP-9 secretion along culture (P < .001). MMP-2 secretion was unaffected by PRL. The relative basal expression of IL-1ß mRNA (1.2 ± 0.87) was reduced by 80% in the presence of PRL after 32 hours of incubation of the membranes (P = .001). The expression of the TNF-α mRNA was not modified by the presence of PRL (0.06 ± 0.01) compared with the basal expression levels (0.05 ± 0.01). MMP-9 mRNA basal expression (0.018 ± 0.008) was significantly reduced (P = .001) in the presence of PRL after 32 hours (0.002 ± 0.0005). CONCLUSION: PRL may be a potential candidate as a key signal controlling the expression of signals related to the proinflammatory reaction associated with human labor.


Assuntos
Membranas Extraembrionárias/metabolismo , Interleucina-1beta/metabolismo , Trabalho de Parto/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Prolactina/metabolismo , Nascimento a Termo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas In Vitro , Gravidez , Reação em Cadeia da Polimerase em Tempo Real
5.
J Biomed Mater Res B Appl Biomater ; 109(8): 1216-1229, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33354857

RESUMO

The aim of this study was to perform a systematic review on the clinical applications where chorion membrane (CM) and amnion/chorion membrane (ACM) were used for oral tissue regeneration procedures. Selection of articles was carried out by two evaluators in Pubmed and Scopus databases, and Outcomes (PICO) method was used to select the relevant articles. Clinical studies reporting the use of CM or ACM for oral soft and hard tissue regeneration were included. The research involved 21 studies conducted on 375 human patients. Seven clinical applications of CM and ACM in oral and periodontal surgery were identified: gingival recession treatment, intrabony and furcation defect treatment, alveolar ridge preservation, keratinized gum width augmentation around dental implants, maxillary sinus membrane repair, and large bone defect reconstruction. CM and ACM were compared to negative controls (conventional surgeries without membrane) or to the following materials: collagen membranes, dense polytetrafluoroethylene membranes, platelet-rich fibrin membranes, amnion membranes, and to a bone substitute. Several studies support the use of CM and ACM as an efficient alternative to current techniques for periodontal and oral soft tissue regeneration procedures. However, further studies are necessary to increase the level of evidence and especially to demonstrate their role for bone regeneration.


Assuntos
Âmnio/metabolismo , Córion/metabolismo , Regeneração Óssea , Transplante Ósseo , Defeitos da Furca , Humanos , Membranas Artificiais , Fibrina Rica em Plaquetas/metabolismo , Politetrafluoretileno/química , Procedimentos de Cirurgia Plástica
6.
Obstet Gynecol Clin North Am ; 47(1): 147-162, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32008665

RESUMO

Fetal membranes (FMs) play a role in pregnancy maintenance and promoting parturition at term. The FMs are not just part of the placenta, structurally or functionally. Although attached to the placenta, the amnion has a separate embryologic origin, and the chorion deviates from the placenta by the first month of pregnancy. Other than immune protection, these FM functions are not those of the placenta. FM dysfunction is associated with and may cause adverse pregnancy outcomes. Ongoing research may identify biomarkers for pending preterm premature rupture of the FMs as well as therapeutic agents, to prevent it and resulting preterm birth.


Assuntos
Membranas Extraembrionárias/diagnóstico por imagem , Ruptura Prematura de Membranas Fetais/diagnóstico , Placenta/diagnóstico por imagem , Nascimento Prematuro , Feminino , Humanos , Gravidez
7.
Front Physiol ; 11: 715, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32695021

RESUMO

The placenta and fetal membrane act as a protective barrier throughout pregnancy while maintaining communication and nutrient exchange between the baby and the mother. Disruption of this barrier leads to various pregnancy complications, including preterm birth, which can have lasting negative consequences. Thus, understanding the role of the feto-maternal interface during pregnancy and parturition is vital to advancing basic and clinical research in the field of obstetrics. However, human subject studies are inherently difficult, and appropriate animal models are lacking. Due to these challenges, in vitro cell culture-based studies are most commonly utilized. However, the structure and functions of conventionally used in vitro 2D and 3D models are vastly different from the in vivo environment, making it difficult to fully understand the various factors affecting pregnancy as well as pathways and mechanisms contributing to term and preterm births. This limitation also makes it difficult to develop new therapeutics. The emergence of in vivo-like in vitro models such as organ-on-chip (OOC) platforms can better recapitulate in vivo functions and responses and has the potential to move this field forward significantly. OOC technology brings together two distinct fields, microfluidic engineering and cell/tissue biology, through which diverse human organ structures and functionalities can be built into a laboratory model that better mimics functions and responses of in vivo tissues and organs. In this review, we first provide an overview of the OOC technology, highlight two major designs commonly used in achieving multi-layer co-cultivation of cells, and introduce recently developed OOC models of the feto-maternal interface. As a vital component of this review, we aim to outline progress on the practicality and effectiveness of feto-maternal interface OOC (FM-OOC) models currently used and the advances they have fostered in obstetrics research. Lastly, we provide a perspective on the future basic research and clinical applications of FM-OOC models, and even those that integrate multiple organ systems into a single OOC system that may recreate intrauterine architecture in its entirety, which will accelerate our understanding of feto-maternal communication, induction of preterm labor, drug or toxicant permeability at this vital interface, and development of new therapeutic strategies.

8.
Semin Immunopathol ; 42(4): 431-450, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32785751

RESUMO

Spontaneous preterm birth (PTB) and preterm pre-labor rupture of the membranes (pPROM) are major pregnancy complications. Although PTB and pPROM have common etiologies, they arise from distinct pathophysiologic pathways. Inflammation is a common underlying mechanism in both conditions. Balanced inflammation is required for fetoplacental growth; however, overwhelming inflammation (physiologic at term and pathologic at preterm) can lead to term and preterm parturition. A lack of effective strategies to control inflammation and reduce the risk of PTB and pPROM suggests that there are several modes of the generation of inflammation which may be dependent on the type of uterine tissue. The avascular fetal membrane (amniochorion), which provides structure, support, and protection to the intrauterine cavity, is one of the key contributors of inflammation. Localized membrane inflammation helps tissue remodeling during pregnancy. Two unique mechanisms that generate balanced inflammation are the progressive development of senescence (aging) and cyclic cellular transitions: epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET). The intrauterine build-up of oxidative stress at term or in response to risk factors (preterm) can accelerate senescence and promote a terminal state of EMT, resulting in the accumulation of inflammation. Inflammation degrades the matrix and destabilizes membrane function. Inflammatory mediators from damaged membranes are propagated via extracellular vesicles (EV) to maternal uterine tissues and transition quiescent maternal uterine tissues into an active state of labor. Membrane inflammation and its propagation are fetal signals that may promote parturition. This review summarizes the mechanisms of fetal membrane cellular senescence, transitions, and the generation of inflammation that contributes to term and preterm parturitions.


Assuntos
Ruptura Prematura de Membranas Fetais , Nascimento Prematuro , Membranas Extraembrionárias , Feminino , Ruptura Prematura de Membranas Fetais/etiologia , Humanos , Recém-Nascido , Inflamação/etiologia , Mediadores da Inflamação , Gravidez
9.
Obstet Gynecol Sci ; 62(4): 199-211, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31338337

RESUMO

A better understanding of the underlying mechanisms by which signals from the fetus initiate human parturition is required. Our recent findings support the core hypothesis that oxidative stress (OS) and cellular senescence of the fetal membranes (amnion and chorion) trigger human parturition. Fetal membrane cell senescence at term is a natural physiological response to OS that occurs as a result of increased metabolic demands by the maturing fetus. Fetal membrane senescence is affected by the activation of the p38 mitogen activated kinase-mediated pathway. Similarly, various risk factors of preterm labor and premature rupture of the membranes also cause OS-induced senescence. Data suggest that fetal cell senescence causes inflammatory senescence-associated secretory phenotype (SASP) release. Besides SASP, high mobility group box 1 and cell-free fetal telomere fragments translocate from the nucleus to the cytosol in senescent cells, where they represent damage-associated molecular pattern markers (DAMPs). In fetal membranes, both SASPs and DAMPs augment fetal cell senescence and an associated 'sterile' inflammatory reaction. In senescent cells, DAMPs are encapsulated in extracellular vesicles, specifically exosomes, which are 30-150 nm particles, and propagated to distant sites. Exosomes traffic from the fetus to the maternal side and cause labor-associated inflammatory changes in maternal uterine tissues. Thus, fetal membrane senescence and the inflammation generated from this process functions as a paracrine signaling system during parturition. A better understanding of the premature activation of these signals can provide insights into the mechanisms by which fetal signals initiate preterm parturition.

10.
Am J Reprod Immunol ; 80(6): e13047, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30178469

RESUMO

Oxidative stress (OS) plays a role in uterine tissue remodeling during pregnancy and parturition. While p38 MAPK is an OS-response kinase, a precise functional role is unknown. Therefore, we conducted a systematic review of literature on p38 MAPK expression, activation, and function in reproductive tissues throughout pregnancy and parturition, published between January 1980 and August 2017, using four electronic databases (Web of Science, PubMed, Medline, and CoCHRANE). We identified 418 reports; 108 were selected for full-text evaluation and 74 were included in final review. p38 MAPK was investigated using feto-maternal primary or immortalized cells, tissue explants, and animal models. Western blot was most commonly used to report phosphorylated (active) p38 MAPK. Human placenta (27), chorioamniotic membranes (14), myometrium (13), decidua (8), and cervix (1) were the studied tissues. p38 MAPK's functions were tissue and gestational age dependent. Isoform specificity was hardly reported. p38 MAPK activity was induced by ROS or proinflammatory cytokines to promote cell signaling linked to cell fate, primed uterus, ripened cervix, and proinflammatory cytokine/chemokine production. In 35 years, reports on p38 MAPK's role during pregnancy and parturition are scarce and current literature is insufficient to provide a comprehensive description of p38 MAPK's mechanistic role during pregnancy and parturition.


Assuntos
Genitália Feminina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Inflamação , Estresse Oxidativo , Parto , Gravidez , Reprodução , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética
11.
Am J Reprod Immunol ; 80(4): e12999, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29911323

RESUMO

PROBLEM: Oxidative stress (OS)-induced, p38 mitogen-activated protein kinase (p38MAPK)-mediated chorioamniotic senescence and inflammation (senescence-associated secretory phenotype [SASP]) are associated with parturition. In response to OS-inducing risk factors, premature senescence contributes to preterm premature rupture of the membranes (pPROM) and spontaneous preterm birth (PTB). We determined the effect of simvastatin, rosuvastatin, and progesterone in downregulating p38MAPK-mediated senescence and SASP. METHOD OF STUDY: Normal term, not-in-labor fetal membranes (n = 8) were exposed to cigarette smoke extract (CSE: OS inducer) alone or combined with simvastatin (100 and 200 ng/mL), rosuvastatin (100 and 200 ng/mL), and progesterone (10-6  mol/L). p38MAPK expression changes were studied by Western blot, senescence was determined by senescence-associated ß-Galactosidase (SA-ß-Gal) staining, and multiplex analysis determined changes associated with 4 SASP markers (IL-8, IL-10, TNF-α, and GM-CSF). A pairwise comparison between groups was conducted by ANOVA. RESULTS: Compared to untreated controls, CSE induced p38MAPK-mediated senescence and SASP. CSE cotreatment with simvastatin and rosuvastatin significantly reduced p38MAPK activation, senescence (decrease in SA-ß-Gal) and SASP markers, GM-CSF, and TNF, but not IL-8, while increasing anti-inflammatory IL-10 in a dose-dependent manner. Cotreatment of CSE and progesterone had no effect on reducing p38MAPK activation, senescence, or SASP. CONCLUSION: Both simvastatin and rosuvastatin downregulated OS-induced p38MAPK activation, senescence, and SASP, while rosuvastatin showed a pronounced effect. Progesterone did not reduce OS-induced fetal membrane senescence and SASP. Simvastatin or rosuvastatin may reduce the incidences of OS-associated PTB and pPROM by preventing premature senescence and SASP.


Assuntos
Senescência Celular/efeitos dos fármacos , Membranas Extraembrionárias/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Progesterona/farmacologia , Rosuvastatina Cálcica/farmacologia , Sinvastatina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adulto , Membrana Corioalantoide/citologia , Membrana Corioalantoide/metabolismo , Feminino , Humanos , Imidazóis/farmacologia , Estresse Oxidativo/fisiologia , Gravidez , Piridinas/farmacologia , Fumaça/análise , Adulto Jovem
12.
J Reprod Immunol ; 123: 3-11, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28858636

RESUMO

Term labor in humans is associated with increased oxidative stress (OS) -induced senescence and damages to amnion epithelial cells (AECs). Senescent fetal cells release alarmin high-mobility group box 1 (HMGB1) and cell-free fetal telomere fragments (cffTF) which can be carried by exosomes to other uterine tissues to produce parturition-associated inflammatory changes. This study characterized AEC-derived exosomes under normal and OS conditions and their packaging of HMGB1 and cffTF. Primary AECs were treated with either standard media or oxidative stress-induced media (exposure to cigarette smoke extract for 48h). Senescence was determined, and exosomes were isolated and characterized. To colocalize HMGB1 and cffTF in amnion exosomes, immunofluorescent staining and in situ hybridization were performed, followed by confocal microscopy. Next generation sequencing (NGS) determined exosomal cffTF and other cell-free amnion cell DNA specificity. Regardless of condition, primary AECs produce exosomes with a classic size, shape, and markers. OS and senescence caused the translocation of HMGB1 and cffTF from AECs' nuclei to cytoplasm compared to untreated cells, which was inhibited by antioxidant N-acetyl cysteine (NAC). Linescans confirmed colocalization of HMGB1 and cffTF in exosomes were higher in the cytoplasm after CSE treatment compared to untreated AECs. NGS determined that besides cffTF, AEC exosomes also carry genomic and mitochondrial DNA, regardless of growth conditions. Sterile inflammatory markers HMGB1 and cffTF from senescent fetal cells are packaged inside exosomes. We postulate that this exosomal cargo can act as a fetal signal at term and can cause labor-associated changes in neighboring tissues.


Assuntos
Alarminas/metabolismo , Âmnio/fisiologia , Células Epiteliais/patologia , Exossomos/metabolismo , Proteína HMGB1/metabolismo , Inflamação/metabolismo , Telômero/metabolismo , Células Cultivadas , Senescência Celular , Fumar Cigarros/efeitos adversos , Meios de Cultivo Condicionados/efeitos adversos , Exossomos/patologia , Feminino , Humanos , Inflamação/genética , Trabalho de Parto , Estresse Oxidativo , Parto , Gravidez , Cultura Primária de Células
13.
Semin Perinatol ; 41(7): 409-419, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28807394

RESUMO

Preterm prelabor rupture of the membranes (pPROM) remains a significant obstetric problem that affects 3-4% of all pregnancies and precedes 40-50% of all preterm births. pPROM arises from complex, multifaceted pathways. In this review, we summarize some old concepts and introduce some novel theories related to pPROM pathophysiology. Specifically, we introduce the concept that pPROM is a disease of the fetal membranes where inflammation-oxidative stress axis plays a major role in producing pathways that can lead to membrane weakening through a variety of processes. In addition, we report microfractures in fetal membranes that are likely sites of tissue remodeling during gestation; however, increase in number and morphometry (width and depth) of these microfractures in pPROM membranes suggests reduced remodeling capacity of membranes. Microfractures can act as channels for amniotic fluid leak, and inflammatory cell and microbial migration. Further studies on senescence activation and microfracture formation and their role in maintaining membrane homeostasis are needed to fill the knowledge gaps in our understanding of pPROM as well as provide better screening (biomarker and imaging based) tools for predicting women at high risk for pPROM and subsequent preterm birth.


Assuntos
Ruptura Prematura de Membranas Fetais/fisiopatologia , Inflamação/fisiopatologia , Nascimento Prematuro/fisiopatologia , Saúde Reprodutiva , Biomarcadores/metabolismo , Membranas Extraembrionárias , Feminino , Ruptura Prematura de Membranas Fetais/metabolismo , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Inflamação/metabolismo , Estresse Oxidativo , Gravidez
14.
Placenta ; 53: 66-75, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28487023

RESUMO

INTRODUCTION: Remodeling of human placental membranes (amniochorionic or fetalmembrane) throughout gestation, a necessity to accommodate increasing uterine volume, involves continuous alterations (replacement of cells and remodeling of extracellular matrix). Methodologic limitations have obscured microscopic determination of cellular and layer-level alterations. This study used a combination of advanced imaging by multiphoton autofluorescence microscopy (MPAM) and second harmonic generation (SHG) microscopy along with tissue optical clearing to characterize the 3Dimensional multilayer organization of placental membranes. METHODS: Placental membranes biopsies (6 mm) collected from term, not-in-labor cesarean deliveries (n = 7) were fixed in 10% formalin (native) or treated with 2,2'-thiodiethanol to render them transparent for deeper imaging. Native and cleared tissues were imaged using MPAM (cellular autofluorescence) and SHG (fibrillar collagen). Depth z-stacks captured the amnion epithelium, underlying matrix layers, and in the cleared biopsies, the decidua layer. RESULTS: MPAM and SHG revealed fetal membrane epithelial topography and collagen organization in multiple matrix layers. Term amnion layers showed epithelial shedding and gaps. Optical clearing provided full-depth imaging with improved visualization of collagen structure, mesenchymal cells in extracellular matrix layers, and decidua morphology. Layer thicknesses measured by imaging corroborated with histology. Mosaic tiling of MPAM/SHG image stacks allowed large area visualization of entire biopsies. CONCLUSION: MPAM-SHG microscopy allowed for study of this multi-layered tissue and revealed shedding, gap formation, and other structural changes. This approach could be used to study structural changes associated with membranes as well as other uterine tissues to better understand events in normal and abnormal parturition.


Assuntos
Membranas Extraembrionárias/diagnóstico por imagem , Microscopia de Fluorescência por Excitação Multifotônica , Placenta/diagnóstico por imagem , Microscopia de Geração do Segundo Harmônico , Matriz Extracelular , Membranas Extraembrionárias/anatomia & histologia , Feminino , Técnicas de Preparação Histocitológica , Humanos , Imageamento Tridimensional , Placenta/anatomia & histologia , Gravidez
16.
Placenta ; 44: 1-5, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27452431

RESUMO

Various endocrine, immune, and mechanical factors produced by feto-maternal compartments at term increase intrauterine inflammatory loads to induce labor. The role of fetal (placental) membranes (amniochorion) as providers of parturition signals has not been well investigated. Fetal membranes line the intrauterine cavity and grow with and protect the fetus. Fetal membranes exist as an entity between the mother and fetus and perform unique functions during pregnancy. Membranes undergo a telomere-dependent p38 MAPK-induced senescence and demonstrate a decline in functional and mechanical abilities at term, showing signs of aging. Fetal membrane senescence is also allied with completion of fetal maturation at term as the fetus readies for delivery, which may also indicate the end of independent life and longevity of fetal membranes as their functional role concludes. Fetal membrane senescence is accelerated at term because of oxidative stress and increased stretching. Senescent fetal membranes cells produce senescence-associated secretory phenotype (SASP-inflammation) and also release proinflammatory damage-associated molecular patterns (DAMPs), namely HMGB1 and cell-free fetal telomere fragments. In a feedback loop, SASP and DAMPs increase senescence and enhance the inflammatory load to promote labor. Membranes increase the inflammatory load to disrupt homeostatic balance to transition quiescent uterine tissues toward a labor phenotype. Therefore, along with other well-described labor-promoting signals, senescent fetal membranes may also contribute to human term parturition.


Assuntos
Membranas Extraembrionárias/metabolismo , Inflamação/metabolismo , Trabalho de Parto/metabolismo , Parto/metabolismo , Senescência Celular/fisiologia , Feminino , Humanos , Gravidez
17.
Am J Reprod Immunol ; 71(1): 86-93, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24286217

RESUMO

PROBLEM: Human parturition is associated with an intrauterine pro-inflammatory environment in the choriodecidua. Evidence that some mediators of this signaling cascade also elicit responses leading to labor prompted us to characterize the cellular sources of these mediators in the human choriodecidua. METHOD OF STUDY: Leukocyte-enriched preparations from human choriodecidua (ChL) and intervillous placental blood leukocytes (PL) were maintained in culture. Secretions of inflammatory cytokines, chemokines, and MMP-9 were documented. Leukocyte phenotype of ChL and PL was determined by flow cytometry using specific fluorochrome-conjugated antibodies. RESULTS AND CONCLUSIONS: ChL showed a distinct pro-inflammatory secretion pattern of cytokines and chemokines when compared with PL, including higher amounts of TNF-α and IL-6, and decreased secretions of IL-4 and IL-1ra. ChL also secreted more MIP-1α and MCP-1 and MMP-9 than PL. No significant differences were found in leukocytes subsets between compartments. Based on our findings, we propose that ChL isolated from fetal membranes at term are functionally different from PL and may collaborate to modulate the microenvironment linked to induction and progression of human labor.


Assuntos
Córion/imunologia , Decídua/imunologia , Leucócitos Mononucleares/imunologia , Subpopulações de Linfócitos/imunologia , Placenta/imunologia , Células Cultivadas , Microambiente Celular , Citocinas/metabolismo , Feminino , Humanos , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Gravidez , Nascimento a Termo/imunologia
18.
Eur J Obstet Gynecol Reprod Biol ; 171(1): 12-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24125907

RESUMO

OBJECTIVES: To determine whether histologic chorioamnionitis is associated with changes in gene expression of TLR-1, -2, -4 and -6, and to describe the localization of these receptors in fetal membranes. STUDY DESIGN: A total of 135 amniochorion membranes with or without histologic chorioamnionitis from preterm or term deliveries were included. Fragments of membranes were submitted to total RNA extraction. RNA was reverse transcribed and the quantification of TLRs expression measured by real time PCR. RESULTS: All amniochorion membranes expressed TLR-1 and TLR-4, whereas 99.1% of membranes expressed TLR-2 and 77.4% expressed TLR-6. TLR-1 and TLR-2 expressions were significantly higher in membranes with histologic chorioamnionitis as compared to membranes without chorioamnionitis in preterm pregnancies (p=0.003 and p<0.001, respectively). Among the membranes of term pregnancies there were no differences in the expressions of such receptors regardless of inflammatory status. Regarding TLR-4 and TLR-6 expression, there was no difference among membranes with or without histologic chorioamnionitis, regardless gestational age at delivery. TLR-1, TLR-2, TLR-4 and TLR-6 expressions were observed in amniotic epithelial, chorionic and decidual cells. CONCLUSION: Amniochorion membranes express TLR-1, TLR-2, TLR-4 and TLR-6 and increased expression of TLR-1 and TLR-2 is related to the presence of histologic chorioamnionitis in preterm pregnancies. This study provides further evidence that amniochorion membranes act as a mechanical barrier to microorganisms and as components of the innate immune system.


Assuntos
Corioamnionite/genética , Receptores Toll-Like/biossíntese , Adulto , Âmnio/metabolismo , Corioamnionite/metabolismo , Córion/metabolismo , Estudos Transversais , Membranas Extraembrionárias/metabolismo , Feminino , Expressão Gênica , Humanos , Trabalho de Parto Prematuro/metabolismo , Gravidez , Nascimento Prematuro/fisiopatologia , Receptor 1 Toll-Like/biossíntese , Receptor 2 Toll-Like/biossíntese , Receptor 4 Toll-Like/biossíntese , Receptor 6 Toll-Like/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA