Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(25): e2400601121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38861608

RESUMEN

The process of human parturition involves inflammation at the interface where fetal chorion trophoblast cells interact with maternal decidual stromal (DS) cells and maternal immune cells in the decidua (endometrium of pregnancy). This study tested the hypothesis that inflammation at the chorion-decidua interface (CDI) induces labor by negating the capacity for progesterone (P4) to block labor and that this is mediated by inactivation of P4 in DS cells by aldo-keto reductase family 1 member C1 (AKR1C1). In human, Rhesus macaque, and mouse CDI, AKR1C1 expression increased in association with term and preterm labor. In a human DS cell line and in explant cultures of term human fetal membranes containing the CDI, the prolabor inflammatory cytokine, interleukin-1ß (IL-1ß), and media conditioned by LPS-stimulated macrophages increased AKR1C1 expression and coordinately reduced nuclear P4 levels and P4 responsiveness. Loss of P4 responsiveness was overcome by inhibition of AKR1C1 activity, inhibition of AKR1C1 expression, and bypassing AKR1C1 activity with a P4 analog that is not metabolized by AKR1C1. Increased P4 activity in response to AKR1C1 inhibition was prevented by the P4 receptor antagonist RU486. Pharmacologic inhibition of AKR1C1 activity prevented parturition in a mouse model of inflammation-induced preterm parturition. The data suggest that inflammatory stimuli at the CDI drive labor by inducing AKR1C1-mediated P4 inactivation in DS cells and that inhibiting and/or bypassing of AKR1C1-mediated P4 inactivation is a plausible therapeutic strategy to mitigate the risk of inflammation-associated preterm birth.


Asunto(s)
20-Hidroxiesteroide Deshidrogenasas , Decidua , Inflamación , Macaca mulatta , Parto , Progesterona , Células del Estroma , Femenino , Animales , Progesterona/metabolismo , Progesterona/farmacología , Decidua/metabolismo , Humanos , Ratones , Células del Estroma/metabolismo , Embarazo , Inflamación/metabolismo , 20-Hidroxiesteroide Deshidrogenasas/metabolismo , 20-Hidroxiesteroide Deshidrogenasas/genética , Interleucina-1beta/metabolismo , Corion/metabolismo
2.
J Neuroinflammation ; 21(1): 16, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200558

RESUMEN

BACKGROUND: Preterm birth is often associated with chorioamnionitis and leads to increased risk of neurodevelopmental disorders, such as autism. Preterm birth can lead to cerebellar underdevelopment, but the mechanisms of disrupted cerebellar development in preterm infants are not well understood. The cerebellum is consistently affected in people with autism spectrum disorders, showing reduction of Purkinje cells, decreased cerebellar grey matter, and altered connectivity. METHODS: Preterm rhesus macaque fetuses were exposed to intra-amniotic LPS (1 mg, E. coli O55:B5) at 127 days (80%) gestation and delivered by c-section 5 days after injections. Maternal and fetal plasma were sampled for cytokine measurements. Chorio-decidua was analyzed for immune cell populations by flow cytometry. Fetal cerebellum was sampled for histology and molecular analysis by single-nuclei RNA-sequencing (snRNA-seq) on a 10× chromium platform. snRNA-seq data were analyzed for differences in cell populations, cell-type specific gene expression, and inferred cellular communications. RESULTS: We leveraged snRNA-seq of the cerebellum in a clinically relevant rhesus macaque model of chorioamnionitis and preterm birth, to show that chorioamnionitis leads to Purkinje cell loss and disrupted maturation of granule cells and oligodendrocytes in the fetal cerebellum at late gestation. Purkinje cell loss is accompanied by decreased sonic hedgehog signaling from Purkinje cells to granule cells, which show an accelerated maturation, and to oligodendrocytes, which show accelerated maturation from pre-oligodendrocytes into myelinating oligodendrocytes. CONCLUSION: These findings suggest a role of chorioamnionitis on disrupted cerebellar maturation associated with preterm birth and on the pathogenesis of neurodevelopmental disorders among preterm infants.


Asunto(s)
Corioamnionitis , Nacimiento Prematuro , Recién Nacido , Femenino , Lactante , Animales , Humanos , Embarazo , Proteínas Hedgehog , Macaca mulatta , Escherichia coli , Recien Nacido Prematuro , Cerebelo , ARN Nuclear Pequeño
3.
PLoS Biol ; 19(9): e3001385, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34495952

RESUMEN

Intrauterine infection/inflammation (IUI) is a major contributor to preterm labor (PTL). However, IUI does not invariably cause PTL. We hypothesized that quantitative and qualitative differences in immune response exist in subjects with or without PTL. To define the triggers for PTL, we developed rhesus macaque models of IUI driven by lipopolysaccharide (LPS) or live Escherichia coli. PTL did not occur in LPS challenged rhesus macaques, while E. coli-infected animals frequently delivered preterm. Although LPS and live E. coli both caused immune cell infiltration, E. coli-infected animals showed higher levels of inflammatory mediators, particularly interleukin 6 (IL-6) and prostaglandins, in the chorioamnion-decidua and amniotic fluid (AF). Neutrophil infiltration in the chorio-decidua was a common feature to both LPS and E. coli. However, neutrophilic infiltration and IL6 and PTGS2 expression in the amnion was specifically induced by live E. coli. RNA sequencing (RNA-seq) analysis of fetal membranes revealed that specific pathways involved in augmentation of inflammation including type I interferon (IFN) response, chemotaxis, sumoylation, and iron homeostasis were up-regulated in the E. coli group compared to the LPS group. Our data suggest that the intensity of the host immune response to IUI may determine susceptibility to PTL.


Asunto(s)
Inmunidad , Trabajo de Parto Prematuro/patología , Complicaciones del Embarazo/inmunología , Animales , Modelos Animales de Enfermedad , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/inmunología , Femenino , Inflamación , Lipopolisacáridos/toxicidad , Macaca mulatta , Embarazo
4.
J Immunol ; 204(10): 2651-2660, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32238461

RESUMEN

Preterm birth (PTB) is a major cause of neonatal mortality and morbidity, often triggered by chorioamnionitis or intrauterine inflammation (IUI) with or without infection. Recently, there has been a strong association of IL-1 with PTB. We hypothesized that IL-1R-associated kinase 1 (IRAK1), a key signaling mediator in the TLR/IL-1 pathway, plays a critical role in PTB. In human fetal membranes (FM) collected immediately after birth from women delivering preterm, p-IRAK1 was significantly increased in all the layers of FM with chorioamnionitis, compared with no-chorioamnionitis subjects. In a preterm rhesus macaque model of IUI given intra-amniotic LPS, induction of p-IRAK1 and downstream proinflammatory signaling mediators were seen in the FM. In a C57BL/6J wild-type PTB mouse model of IUI given intrauterine LPS, an IRAK1 inhibitor significantly decreased PTB and increased live birth in a dose-dependent manner. Furthermore, IRAK1 knockout mice were protected from LPS-induced PTB, which was seen in wild-type controls. Activation of IRAK1 was maintained by K63-mediated ubiquitination in preterm FM of humans with chorioamnionitis and rhesus and mouse IUI models. Mechanistically, IRAK1 induced PTB in the mouse model of IUI by upregulating expression of COX-2. Thus, our data from human, rhesus, and mouse demonstrates a critical role IRAK1 in IUI and inflammation-associated PTB and suggest it as potential therapeutic target in IUI-induced PTB.


Asunto(s)
Membranas Extraembrionarias/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Nacimiento Prematuro/metabolismo , Útero/inmunología , Adulto , Animales , Corioamnionitis , Modelos Animales de Enfermedad , Membranas Extraembrionarias/patología , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Lipopolisacáridos/inmunología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Nacimiento Prematuro/inmunología , Adulto Joven
5.
J Immunol ; 196(9): 3706-15, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036917

RESUMEN

Chorioamnionitis is associated with preterm labor and fetal inflammatory response syndrome (FIRS), causing fetal organ injury and morbidity, particularly in extremely premature infants. However, the effects of inflammation on the fetal immune system remain poorly understood, due to the difficulty of studying immune development in infants. Therefore, we used the model of intra-amniotic LPS administered at ∼80% gestation in rhesus monkeys to cause chorioamnionitis and FIRS that is similar in human pathology. Importantly, the frequency of IL-17(+) and IL-22(+) CD4(+) T cells increased in the spleen of LPS-exposed fetuses, whereas regulatory T cell (Treg) frequency decreased. These changes persisted for at least 48 h. Notably, Th17 cytokines were predominantly expressed by FOXP3(+)CD4(+) T cells and not by their FOXP3(-) counterparts. Bifunctional IL-17(+)FOXP3(+) exhibited a phenotype of inflammatory Tregs (RORc(High/+), Helios(Low/-), IL-2(+), IFN-γ(+), and IL-8(+)) compared with typical FOXP3(+) cells. Diminished splenic Treg frequency in LPS-exposed fetuses was associated with inadequate Treg generation in the thymus. Mechanistically, the emergence of inflammatory Tregs was largely dependent on IL-1 signaling. However, blockage of IL-1R signaling did not abolish the deleterious effects of LPS on Treg frequency in the thymus or spleen. Collectively, we demonstrate that a prenatal inflammatory environment leads to inadequate Treg generation in the thymus with a switch of splenic Tregs toward an inflammatory phenotype. Both processes likely contribute to the pathogenesis of chorioamnionitis. Approaches to manipulate Treg numbers and function could thus be useful therapeutically to alleviate FIRS in preterm infants.


Asunto(s)
Corioamnionitis/inmunología , Inmunoterapia/tendencias , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Interleucina-1/metabolismo , Trabajo de Parto Prematuro/inmunología , Linfocitos T Reguladores/inmunología , Animales , Corioamnionitis/terapia , Modelos Animales de Enfermedad , Femenino , Feto , Factores de Transcripción Forkhead/metabolismo , Humanos , Lipopolisacáridos/inmunología , Macaca mulatta , Trabajo de Parto Prematuro/terapia , Embarazo , Transducción de Señal
6.
J Infect Dis ; 214(10): 1597-1604, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27601620

RESUMEN

BACKGROUND: Although Ureaplasma species are the most common organisms associated with prematurity, their effects on the maternal and fetal immune system remain poorly characterized. METHODS: Rhesus macaque dams at approximately 80% gestation were injected intra-amniotically with 107 colony-forming units of Ureaplasma parvum or saline (control). Fetuses were delivered surgically 3 or 7 days later. We performed comprehensive assessments of inflammation and immune effects in multiple fetal and maternal tissues. RESULTS: Although U. parvum grew well in amniotic fluid, there was minimal chorioamnionitis. U. parvum colonized the fetal lung, but fetal systemic microbial invasion was limited. Fetal lung inflammation was mild, with elevations in CXCL8, tumor necrosis factor (TNF) α, and CCL2 levels in alveolar washes at day 7. Inflammation was not detected in the fetal brain. Significantly, U. parvum decreased regulatory T cells (Tregs) and activated interferon γ production in these Tregs in the fetus. It was detected in uterine tissue by day 7 and induced mild inflammation and increased expression of connexin 43, a gap junction protein involved with labor. CONCLUSIONS: U. parvum colonized the amniotic fluid and caused uterine inflammation, but without overt chorioamnionitis. It caused mild fetal lung inflammation but had a more profound effect on the fetal immune system, decreasing Tregs and polarizing them toward a T-helper 1 phenotype.


Asunto(s)
Líquido Amniótico/microbiología , Corioamnionitis/patología , Endometritis/patología , Enfermedades Fetales/patología , Infecciones por Ureaplasma/patología , Ureaplasma/inmunología , Animales , Corioamnionitis/inmunología , Modelos Animales de Enfermedad , Endometritis/inmunología , Femenino , Enfermedades Fetales/inmunología , Macaca mulatta , Embarazo , Ureaplasma/aislamiento & purificación , Infecciones por Ureaplasma/inmunología
7.
Immunology ; 144(4): 661-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25351513

RESUMEN

CD91 is a scavenger receptor expressed by different immune cells and its ligands defensins have been demonstrated to contribute to immune responses against infections and tumours. We previously demonstrated that CD91 is expressed on human monocyte-derived dendritic cells (moDCs) and that human defensins stimulate in vitro the activation of these cells. In this study, we observed that CD91 is expressed at different levels on two distinct moDC subsets: CD91(dim) and CD91(bright) moDCs. Although CD91(bright) moDCs represented a small proportion of total moDCs, this subset showed higher levels of activation and maturation markers compared with CD91(dim) moDCs. The frequency of CD91(bright) moDCs increased by ~ 50% after in vitro stimulation with recombinant human neutrophil peptide-1 (rHNP-1) and recombinant human ß defensin-1 (rHBD-1), while lipopolysaccharide (LPS) stimulation decreased it by ~ 35%. Both defensins up-regulated moDC expression of CD80, CD40, CD83 and HLA-DR, although to a lower extent compared with LPS. Notably, upon culture with rHNP-1 and rHBD-1, CD91(bright) moDCs maintained their higher activation/maturation status, whereas this was lost upon culture with LPS. Our findings suggest that defensins promote the differentiation into activated CD91(bright) DCs and may encourage the exploitation of the CD91/defensins axis as a novel therapeutic strategy to potentiate antimicrobial and anti-tumour immune response.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , alfa-Defensinas/farmacología , beta-Defensinas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/clasificación , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Humanos , Lipopolisacáridos/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/inmunología , Fenotipo , Proteínas Recombinantes/farmacología , Factores de Tiempo , Regulación hacia Arriba
8.
Biol Reprod ; 92(2): 56, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25537373

RESUMEN

Chorioamnionitis, an infection/inflammation of the fetomaternal membranes, is frequently associated with preterm delivery. The mechanisms of inflammation in chorioamnionitis are poorly understood. We hypothesized that neutrophils recruited to the decidua would be the major producers of proinflammatory cytokines. We injected intra-amniotic (IA) interleukin 1beta (IL-1beta) at ∼80% gestation in rhesus macaque monkeys, Macaca mulatta, delivered the fetuses surgically 24 h or 72 h after IA injections, and investigated the role of immune cells in the chorion-amnion decidua. IA IL-1beta induced a robust infiltration of neutrophils and significant increases of proinflammatory cytokines in the chorioamnion decidua at 24 h after exposure, with a subsequent decrease at 72 h. Neutrophils in the decidua were the major source of tumor necrosis factor alpha (TNFalpha) and IL-8. Interestingly, IA IL-1beta also induced a significant increase in anti-inflammatory indoleamine 2,3-dioxygenase (IDO) expression in the decidua neutrophils. The frequency of regulatory T cells (Tregs) and FOXP3 mRNA expression in the decidua did not change after IA IL-1beta injection. Collectively, our data demonstrate that in this model of sterile chorioamnionitis, the decidua neutrophils cause the inflammation in the gestational tissues but may also act as regulators to dampen the inflammation. These results help to understand the contribution of neutrophils to the pathogenesis of chorioamnionitis-induced preterm labor.


Asunto(s)
Decidua/efectos de los fármacos , Interleucina-1beta/farmacología , Interleucina-8/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Decidua/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Macaca mulatta , Infiltración Neutrófila/fisiología , Embarazo , Nacimiento Prematuro/metabolismo , Linfocitos T Reguladores/metabolismo
9.
J Immunol ; 191(3): 1102-9, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23794628

RESUMEN

Very low birth weight preterm newborns are susceptible to the development of debilitating inflammatory diseases, many of which are associated with chorioamnionitis. To define the effects of chorioamnionitis on the fetal immune system, IL-1ß was administered intra-amniotically at ~80% gestation in rhesus monkeys. IL-1ß caused histological chorioamnionitis, as well as lung inflammation (infiltration of neutrophils or monocytes in the fetal airways). There were large increases in multiple proinflammatory cytokine mRNAs in the lungs at 24 h postadministration, which remained elevated relative to controls at 72 h. Intra-amniotic IL-1ß also induced the sustained expression of the surfactant proteins in the lungs. Importantly, IL-1ß significantly altered the balance between inflammatory and regulatory T cells. Twenty-four hours after IL-1ß injection, the frequency of CD3(+)CD4(+)FOXP3(+) T cells was decreased in lymphoid organs. In contrast, IL-17A-producing cells (CD3(+)CD4(+), CD3(+)CD4(-), and CD3(-)CD4(-) subsets) were increased in lymphoid organs. The frequency of IFN-γ-expressing cells did not change. In this model of a single exposure to an inflammatory trigger, CD3(+)CD4(+)FOXP3(+) cells rebounded quickly, and their frequency was increased at 72 h compared with controls. IL-17 expression was also transient. Interestingly, the T cell profile alteration was confined to the lymphoid organs and not to circulating fetal T cells. Together, these results suggest that the chorioamnionitis-induced IL-1/IL-17 axis is involved in the severe inflammation that can develop in preterm newborns. Boosting regulatory T cells and/or controlling IL-17 may provide a means to ameliorate these abnormalities.


Asunto(s)
Corioamnionitis/inmunología , Inflamación/inmunología , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Linfocitos T Reguladores/inmunología , Líquido Amniótico/química , Líquido Amniótico/citología , Animales , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos , Femenino , Feto/efectos de los fármacos , Feto/metabolismo , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Interleucina-1/metabolismo , Interleucina-1beta/farmacología , Recuento de Linfocitos , Macaca mulatta , Neumonía/inducido químicamente , Neumonía/metabolismo , Embarazo , Surfactantes Pulmonares/metabolismo , ARN Mensajero/genética , Subgrupos de Linfocitos T
10.
Front Immunol ; 15: 1416162, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38895127

RESUMEN

Introduction: IL6 signaling plays an important role in triggering labor and IL6 is an established biomarker of intrauterine infection/inflammation (IUI) driven preterm labor (PTL). The biology of IL6 during IUI at the maternal-fetal interface was investigated in samples from human subjects and non-human primates (NHP). Methods: Pregnant women with histologic chorioamnionitis diagnosed by placenta histology were recruited (n=28 term, n=43 for preterm pregnancies from 26-36 completed weeks of gestation). IUI was induced in Rhesus macaque by intraamniotic injection of lipopolysachharide (LPS, n=23). IL1 signaling was blocked using Anakinra (human IL-1 receptor antagonist, n=13), and Tumor necrosis factor (TNF) signaling was blocked by anti TNF-antibody (Adalimumab n=14). The blockers were given before LPS. All animals including controls (intraamniotic injection of saline n=27), were delivered 16h after LPS/saline exposure at about 80% gestation. Results: IUI induced a robust expression of IL6 mRNAs in the fetal membranes (chorion-amnion-decidua tissue) both in humans (term and preterm) and NHP. The major sources of IL6 mRNA expression were the amnion mesenchymal cells (AMC) and decidua stroma cells. Additionally, during IUI in the NHP, ADAM17 (a protease that cleaves membrane bound IL6 receptor (IL6R) to release a soluble form) and IL6R mRNA increased in the fetal membranes, and the ratio of IL6 and soluble forms of IL6R, gp130 increased in the amniotic fluid signifying upregulation of IL6 trans-signaling. Both IL1 and TNF blockade suppressed LPS-induced IL6 mRNAs in the AMC and variably decreased elements of IL6 trans-signaling. Discussion: These data suggest that IL1 and TNF blockers may be useful anti-inflammatory agents via suppression of IL6 signaling at the maternal-fetal interface.


Asunto(s)
Interleucina-6 , Macaca mulatta , Transducción de Señal , Factor de Necrosis Tumoral alfa , Femenino , Embarazo , Humanos , Animales , Interleucina-6/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Corioamnionitis/inmunología , Corioamnionitis/metabolismo , Corioamnionitis/veterinaria , Lipopolisacáridos/inmunología , Interleucina-1/metabolismo , Adulto , Trabajo de Parto Prematuro/inmunología , Trabajo de Parto Prematuro/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Placenta/metabolismo , Placenta/inmunología
11.
STAR Protoc ; 5(2): 103044, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38678572

RESUMEN

The amnion is a thin layer of fetal origin in contact with the amniotic fluid which plays a key role at the feto-maternal interface during pregnancy. Here, we present a protocol for isolation of human and Rhesusmacaque amnion cells. We describe steps for tissue dissection, cell isolation for flow cytometry analysis, and RNA isolation for RNA sequencing library preparation and analysis. This protocol can provide insights into altered immunological pathways during intrauterine infections to develop new therapeutic strategies. For complete details on the use and execution of this protocol, please refer to Presicce et al.1.


Asunto(s)
Amnios , Separación Celular , Citometría de Flujo , Placenta , Amnios/citología , Humanos , Citometría de Flujo/métodos , Femenino , Embarazo , Animales , Placenta/citología , Separación Celular/métodos , Perfilación de la Expresión Génica/métodos , Transcriptoma/genética , Macaca mulatta
12.
J Virol ; 86(19): 10262-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22811537

RESUMEN

Regulatory T cells (Tregs) play a pivotal role in the maintenance of tolerance as well as in the control of immune activation, particularly during chronic infections. In the setting of HIV infection, the majority of studies have reported an increase in Treg frequency but a decrease in absolute number in all immune compartments of HIV-infected individuals. Several nonexclusive mechanisms have been postulated to explain this preferential Treg accumulation, including peripheral survival, increased proliferation, increased peripheral conversion, and tissue redistribution. The role played by Tregs during HIV infection is still poorly understood, as two opposing hypotheses have been proposed. A detrimental role of Tregs during HIV infection was suggested based on the evidence that Tregs suppress virus-specific immune responses. Conversely, Tregs could be beneficial by limiting immune activation, thus controlling the availability of HIV targets as well as preventing immune-based pathologies. Despite the technical difficulties, getting a better understanding of the mechanisms regulating Treg dynamics remains important, as it will help determine whether we can successfully manipulate Treg function or number to the advantage of the infected host. The aim of this review is thus to discuss the recent findings on Treg homeostasis and function in the setting of HIV infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por VIH/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Linfocitos T Reguladores/virología , Animales , Antivirales/uso terapéutico , Proliferación Celular , Homeostasis , Humanos , Ratones , Modelos Biológicos , Fenotipo , Distribución Tisular , Carga Viral
13.
Cell Rep ; 42(4): 112352, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37027297

RESUMEN

Clinical evidence points to a function for B cell-activating factor (BAFF) in pregnancy. However, direct roles for BAFF-axis members in pregnancy have not been examined. Here, via utility of genetically modified mice, we report that BAFF promotes inflammatory responsiveness and increases susceptibility to inflammation-induced preterm birth (PTB). In contrast, we show that the closely related A proliferation-inducing ligand (APRIL) decreases inflammatory responsiveness and susceptibility to PTB. Known BAFF-axis receptors serve a redundant function in signaling BAFF/APRIL presence in pregnancy. Treatment with anti-BAFF/APRIL monoclonal antibodies or BAFF/APRIL recombinant proteins is sufficient to manipulate susceptibility to PTB. Notably, macrophages at the maternal-fetal interface produce BAFF, while BAFF and APRIL presence divergently shape macrophage gene expression and inflammatory function. Overall, our findings demonstrate that BAFF and APRIL play divergent inflammatory roles in pregnancy and provide therapeutic targets for mitigating risk of inflammation-induced PTB.


Asunto(s)
Nacimiento Prematuro , Animales , Femenino , Ratones , Embarazo , Factor Activador de Células B , Inflamación , Transducción de Señal , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
14.
iScience ; 26(11): 108118, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37953944

RESUMEN

Intrauterine infection/inflammation (IUI) is a frequent complication of pregnancy leading to preterm labor and fetal inflammation. How inflammation is modulated at the maternal-fetal interface is unresolved. We compared transcriptomics of amnion (a fetal tissue in contact with amniotic fluid) in a preterm Rhesus macaque model of IUI induced by lipopolysaccharide with human cohorts of chorioamnionitis. Bulk RNA sequencing (RNA-seq) amnion transcriptomic profiles were remarkably similar in both Rhesus and human subjects and revealed that induction of key labor-mediating genes such as IL1 and IL6 was dependent on nuclear factor κB (NF-κB) signaling and reversed by the anti-tumor necrosis factor (TNF) antibody Adalimumab. Inhibition of collagen biosynthesis by IUI was partially restored by Adalimumab. Interestingly, single-cell transcriptomics, flow cytometry, and immunohistology demonstrated that a subset of amnion mesenchymal cells (AMCs) increase CD14 and other myeloid cell markers during IUI both in the human and Rhesus macaque. Our data suggest that CD14+ AMCs represent activated AMCs at the maternal-fetal interface.

15.
Mucosal Immunol ; 15(4): 730-744, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35314757

RESUMEN

Up to 40% of preterm births are associated with histological chorioamnionitis (HCA), which leads to elevated levels of pro-inflammatory mediators and microbial products in the amniotic fluid, which come in contact with fetal lungs. Yet, fetal pulmonary immune responses to such exposure remain poorly characterized. To address this gap, we used our established HCA model, in which pregnant Rhesus macaques receive intraamniotic (IA) saline or LPS. IA LPS induced a potent and rapid myeloid cell response in fetal lungs, dominated by neutrophils and monocytes/macrophages. Infiltrating and resident myeloid cells exhibited transcriptional profiles consistent with exposure to TLR ligands, as well as cytokines, notably IL-1 and TNFα. Although simultaneous, in vivo blockade of IL-1 and TNFα signaling did not prevent the inflammatory cell recruitment, it blunted the lung overall inflammatory state reducing communication between, and activation of, infiltrating immune cells. Our data indicate that the fetal innate immune system can mount a rapid multi-faceted pulmonary immune response to in utero exposure to inflammation. These data provide mechanistic insights into the association between HCA and the postnatal lung morbidities of the premature infant and highlight therapeutic potential of inflammatory blockade in the fetus.


Asunto(s)
Corioamnionitis , Neumonía , Nacimiento Prematuro , Líquido Amniótico , Animales , Corioamnionitis/patología , Femenino , Humanos , Inflamación , Interleucina-1 , Lipopolisacáridos , Pulmón , Macaca mulatta , Embarazo , Nacimiento Prematuro/patología , Factor de Necrosis Tumoral alfa
16.
Sci Transl Med ; 14(638): eabl8574, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35353543

RESUMEN

Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1ß and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.


Asunto(s)
Corioamnionitis , Nacimiento Prematuro , Animales , Corioamnionitis/inducido químicamente , Corioamnionitis/patología , Femenino , Pulmón/patología , Macaca mulatta , Embarazo , Nacimiento Prematuro/prevención & control , Intercambio Gaseoso Pulmonar
17.
Front Pediatr ; 9: 614209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777863

RESUMEN

Background: Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pro-inflammatory cytokine that is increased in the amniotic fluid in chorioamnionitis and elevated in the fetal lung with endotoxin exposure. Although GM-CSF has a pivotal role in fetal lung development, it stimulates pulmonary macrophages and is associated with the development of bronchopulmonary dysplasia (BPD). How antenatal GM-CSF results in recruitment of lung macrophage leading to BPD needs further elucidation. Hence, we used a transgenic and knock-out mouse model to study the effects of GM-CSF focusing on the fetal lung macrophage. Methods: Using bitransgenic (BTg) mice that conditionally over-expressed pulmonary GM-CSF after doxycycline treatment, and GM-CSF knock-out (KO) mice with no GM-CSF expression, we compared the ontogeny and immunophenotype of lung macrophages in BTg, KO and control mice at various prenatal and postnatal time points using flow cytometry and immunohistology. Results: During fetal life, compared to controls, BTg mice over-expressing pulmonary GM-CSF had increased numbers of lung macrophages that were CD68+ and these were primarily located in the interstitium rather than alveolar spaces. The lung macrophages that accumulated were predominantly CD11b+F4/80+ indicating immature macrophages. Conversely, lung macrophages although markedly reduced, were still present in GM-CSF KO mice. Conclusion: Increased exposure to GM-CSF antenatally, resulted in accumulation of immature macrophages in the fetal lung interstitium. Absence of GM-CSF did not abrogate but delayed the transitioning of interstitial macrophages. Together, these results suggest that other perinatal factors may be involved in modulating the maturation of alveolar macrophages in the developing fetal lung.

18.
Cytometry A ; 77(6): 571-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20162533

RESUMEN

FOXP3 is a key transcription factor expressed by regulatory T cells (Treg cells). However, differences in staining and analysis protocols have led to conflicting results. Moreover, the transient upregulation of FOXP3 that follows activation in non-Treg cells renders the interpretation of FOXP3 data more difficult in humans than in mice. Human peripheral blood mononuclear cells (PBMCs), isolated CD25(-) or CD25(+)CD4(+) T cells were stained with three different anti-FOXP3 clones (PCH101, 206D, and 259D) alone or in combination, and using different permeabilization methods. FOXP3 expression was evaluated following T cell activation by several pathways. Gating based on a population that did not express FOXP3 (such as CD3(-)CD4(-) T cells) allowed for the optimal characterization of Treg cells. The 206D clone detected a lower percentage of cells than PCH101 or 259D. In contrast, 259D stained a population of activated T cells that PCH101 did not. Staining with two clones together consistently increased the proportion of FOXP3(+) cells. However, it is likely that only the double positive cells are Treg cells, as they expressed the highest CD25 and lowest CD127 levels. Our results emphasize that the choice of staining protocol leads to very different results concerning the frequency of Treg cells in humans. A more consistent identification of these cells will improve the knowledge of their biology, particularly during disease processes.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Coloración y Etiquetado/métodos , Biomarcadores/metabolismo , Células Cultivadas , Células Clonales , Factores de Transcripción Forkhead/química , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Estándares de Referencia , Transducción de Señal , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/metabolismo
19.
Front Immunol ; 11: 649, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32373122

RESUMEN

Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.


Asunto(s)
Corioamnionitis/inmunología , Infecciones/inmunología , Vagina/inmunología , Animales , Disbiosis , Femenino , Humanos , Microbiota , Embarazo , Complicaciones Infecciosas del Embarazo , Vagina/microbiología
20.
Front Immunol ; 11: 866, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32528468

RESUMEN

Intra-amniotic (IA) inflammation is associated with significant morbidities for both the mother and the fetus. Prior studies have illustrated many of the effects of IA inflammation on the uterine lining (decidua) and membranous layers of the placenta at the fetal-maternal interface. However, much less is known about the immunological response occurring within the villous placenta. Using a rhesus macaque model of lipopolysaccharide (LPS)-induced IA inflammation, we showed that pregnancy-matched choriodecidua and villi have distinct immunological profiles in rhesus pregnancies. In the choriodecidua, we show that the abundance of neutrophils, multiple populations of antigen-presenting cells, and two populations of natural killer (NK) cells changes with prenatal IA LPS exposure. In contrast, in immune cells within the villous placenta we observed alterations in the abundance of B cells, monocytes, and CD8 T cells. Prior work has illustrated that IA inflammation leads to an increase in tumor necrosis factor alpha (TNFα) at the fetal-maternal interface. In this study, pretreatment with a TNFα blockade partially reversed inflammation in the placental villi. Furthermore, we report that immune cells in the villous placenta sensed LPS during our experimental window, and subsequently activated T cells to produce proinflammatory cytokines. Moreover, this study is the first report of memory T cells in third-trimester non-human primate placental villi and provides evidence that manipulation of immune cells in the villi at the fetal-maternal interface should be considered as a potential therapeutic target for IA inflammation.


Asunto(s)
Corioamnionitis/inmunología , Vellosidades Coriónicas/inmunología , Decidua/inmunología , Leucocitos/inmunología , Activación de Linfocitos , Animales , Biomarcadores/metabolismo , Corioamnionitis/inducido químicamente , Corioamnionitis/tratamiento farmacológico , Corioamnionitis/metabolismo , Vellosidades Coriónicas/efectos de los fármacos , Vellosidades Coriónicas/metabolismo , Decidua/efectos de los fármacos , Decidua/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunofenotipificación , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Lipopolisacáridos , Macaca mulatta , Embarazo , Transducción de Señal , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA