Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Am J Reprod Immunol ; 91(1): e13802, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38282608

RESUMO

Pregnancy is a finely tuned process, with the health and well-being of the developing fetus determined by the metabolic status and dietary intake of the mother. The maternal gut microbiome is remodeled during pregnancy, and this, coupled with the maternal nutrient intake during gestation shapes the production of metabolites that can cross the placenta and affect fetal development. As posited by the Developmental Origins of Health and Disease Hypothesis, such environmental influences can have major effects on the developing organ systems. When occurring at particularly sensitive gestational time points, these developmental programming events can have long lasting effects on offspring adaptation to the postnatal environment, and major health implications later in life. This review will summarize current knowledge on how pregnancy and maternal dietary intake intrinsically and extrinsically modify maternal gut microbiota composition and metabolite production. Further, we will assess how these factors shape the fetal landscape and ultimately contribute to offspring health. DOHaD, fetal development, metabolites, microbiome, nutrition, pregnancy, short-chain fatty acids.


Assuntos
Microbioma Gastrointestinal , Humanos , Gravidez , Feminino , Fenômenos Fisiológicos da Nutrição Pré-Natal , Desenvolvimento Fetal , Placenta/metabolismo , Cuidado Pré-Natal
2.
Acta Physiol (Oxf) ; 240(3): e14092, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38251788

RESUMO

Asthma is the most common chronic disease among children, with more than 300 million cases worldwide. Over the past several decades, asthma incidence has grown, and epidemiological studies identify the modernized lifestyle as playing a strong contributing role in this phenomenon. In particular, lifestyle factors that modify the maternal gut microbiome during pregnancy, or the infant microbiome in early life, can act as developmental programming events which determine health or disease susceptibility later in life. Microbial colonization of the gut begins at birth, and factors such as delivery mode, breastfeeding, diet, antibiotic use, and exposure to environmental bacteria influence the development of the infant microbiome. Colonization of the gut microbiome is crucial for proper immune system development and disruptions to this process can predispose a child to asthma development. Here, we describe the importance of early-life events for shaping immune responses along the gut-lung axis and why they may provide a window of opportunity for asthma prevention.


Assuntos
Asma , Microbioma Gastrointestinal , Recém-Nascido , Criança , Lactente , Feminino , Gravidez , Humanos , Suscetibilidade a Doenças , Estilo de Vida , Pulmão
3.
Eur J Immunol ; 53(10): e2350394, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37431194

RESUMO

Antibiotic use during pregnancy is associated with increased asthma risk in children. Since approximately 25% of women use antibiotics during pregnancy, it is important to identify the pathways involved in this phenomenon. We investigate how mother-to-offspring transfer of antibiotic-induced gut microbial dysbiosis influences immune system development along the gut-lung axis. Using a mouse model of maternal antibiotic exposure during pregnancy, we immunophenotyped offspring in early life and after asthma induction. In early life, prenatal-antibiotic exposed offspring exhibited gut microbial dysbiosis, intestinal inflammation (increased fecal lipocalin-2 and IgA), and dysregulated intestinal ILC3 subtypes. Intestinal barrier dysfunction in the offspring was indicated by a FITC-dextran intestinal permeability assay and circulating lipopolysaccharide. This was accompanied by increased T-helper (Th)17 cell percentages in the offspring's blood and lungs in both early life and after allergy induction. Lung tissue additionally showed increased percentages of RORγt T-regulatory (Treg) cells at both time points. Our investigation of the gut-lung axis identifies early-life gut dysbiosis, intestinal inflammation, and barrier dysfunction as a possible developmental programming event promoting increased expression of RORγt in blood and lung CD4+ T cells that may contribute to increased asthma risk.


Assuntos
Asma , Microbioma Gastrointestinal , Gravidez , Criança , Humanos , Feminino , Antibacterianos/efeitos adversos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Disbiose , Inflamação , Pulmão
4.
Int Immunol ; 35(10): 475-482, 2023 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-37464466

RESUMO

Childhood allergic asthma is associated with a dysbiotic gut microbiome in early life, and maternal perinatal treatment with probiotics is a potential way alter the infant microbiome, which may improve asthma outcomes. This study used a mouse model to examine the effect of maternal supplementation with the probiotic Enterococcus faecalis on faecal short-chain fatty acid (SCFA) concentrations and asthma risk in the offspring. Pregnant/lactating mice were treated daily, from gestation day 6 to postnatal day 21, with an oral suspension of 106, 107 or 108 colony-forming units of a live preparation of the probiotic E. faecalis (Symbioflor®1). At weaning, offspring were subjected to an ovalbumin-induced experimental asthma protocol. Faeces were collected from the mothers and offspring at several different time points to determine SCFA concentrations. It was found that maternal supplementation with E. faecalis did not alter litter size, sex ratio or offspring weight, and was associated with an increase in SCFAs in offspring faeces at weaning and after allergy induction. However, allergic offspring from E. faecalis supplemented mothers showed no difference in asthma severity when compared with allergic offspring from control mothers. In conclusion, although maternal perinatal supplementation with low-dose E. faecalis was associated with increased faecal SCFAs in the offspring, it did not protect against offspring asthma. This is may be because SCFA concentrations were not increased to an immunoprotective level. We recommend that future studies concentrate on probiotic supplementation in high-risk cases, for instance, to repair gut dysbiosis resulting from antibiotic use in pregnant mothers or their infants.


Assuntos
Asma , Hipersensibilidade , Humanos , Gravidez , Lactente , Feminino , Animais , Camundongos , Criança , Enterococcus faecalis , Lactação , Suplementos Nutricionais , Ácidos Graxos Voláteis
6.
Front Immunol ; 13: 999852, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275674

RESUMO

Histamine (HA) is a potent mediator that plays a central role in inflammation and allergy, acting through four G-protein-coupled receptors (i.e. H1-H4). HA is an accepted promoter of type 2 immunity in CD4+ T cells during hypersensitivity. Previously, we demonstrated that HA can promote antigen cross-presentation, inducing the activation of antigen-specific CD8+ T cells in an asthmatic murine model. Non-classical CD8+ T-cell profiles, such as Tc2 or Tc17, are associated with allergic disease persistence and chronicity. In this paper, we focus on the role of the H3 receptor (H3R) and the H4 receptor (H4R) in the development of allergic contact dermatitis. We were able to show that induction of the type 2 profiles associated with interleukin 13 production, both by CD4 and CD8 lymphocytes, depend on the interaction of HA with H3R and H4R. Blocking both receptors using the selective H3/H4 receptor antagonist thioperamide or the selective H4R ligand JNJ777120 reduces the inflammatory response, inducing an immunosuppressive profile associated with the increased proportion of FOXp3+ regulatory T lymphocytes and CD11b+Gr-1+ myeloid suppressor cells. Interestingly, in dendritic cells, only H4R blockade, and not H3R blockade, is capable of modulating most of the inflammatory effects observed in our model.


Assuntos
Dermatite Alérgica de Contato , Histamina , Camundongos , Animais , Receptores Histamínicos H4 , Linfócitos T CD8-Positivos , Ligantes , Interleucina-13 , Receptores Histamínicos , Receptores Acoplados a Proteínas G , Fatores de Transcrição Forkhead
7.
Methods Mol Biol ; 2442: 603-619, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35320548

RESUMO

Galectin-1 (gal-1), a member of a family of evolutionarily conserved glycan-binding proteins, is differentially expressed at the feto-maternal interface and appears to be functionally polyvalent, with a wide range of biological activities. However, the contributions of maternal and/or feto-placental gal-1 to the signaling networks promoting a healthy pregnancy are still being elucidated. This chapter discusses the methods commonly employed to study the maternal or feto-placental contribution of gal-1 during pregnancy in mice. The methods described here can be used to decipher the specific role of each source, e.g., maternal and/or feto-placental derived gal-1 in the orchestration of pregnancy-associated processes.


Assuntos
Galectina 1 , Placenta , Gravidez , Animais , Família , Feminino , Galectina 1/genética , Galectina 1/metabolismo , Camundongos , Placenta/metabolismo , Gravidez/metabolismo , Resultado da Gravidez
8.
Am J Reprod Immunol ; 86(6): e13491, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34363260

RESUMO

PROBLEM: Proper placental development is pivotal to ensure healthy pregnancy outcomes. Among the multiple cellular mechanisms involved in the orchestration of this process, little is known on the role of alternative splicing events in the modulation of trophoblast cell biology. Here, we evaluated the expression of the alternative splicing regulator Rbfox2 in the pre- and post-placentation period in mouse pregnancies in both healthy and pathological settings. METHOD OF STUDY: Immunofluorescence analysis of Rbfox2 expression in mouse implantation sites collected during the pre-placentation period (E5-E7) and post-placentation (E13). RESULTS: We identified a progressive increase of Rbfox2 levels throughout the peri-implantation period with a shift from a cytoplasmatic expression on E5-E6 to a predominantly nuclear expression on E7, together with a prominent expression of this factor in both subcellular compartments of the primitive placenta. Our results further showed that in contrast to healthy gestations, Rbfox2 expression decreased in preeclamptic models during the post-placentation period. Finally, we further demonstrated enhanced expression of Rbfox2 proteins in allogeneic pregnancy compared to syngeneic models. CONCLUSIONS: Our findings uncover a novel role for Rbfox2-controlled splicing events in the modulation of trophoblast function, with potential implications for the pathogenesis of preeclampsia and other pregnancy complications originated from defective placentation.


Assuntos
Regulação da Expressão Gênica , Placentação/genética , Pré-Eclâmpsia/metabolismo , Fatores de Processamento de RNA/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Placenta/metabolismo , Pré-Eclâmpsia/genética , Gravidez , Fatores de Processamento de RNA/genética , Trofoblastos/metabolismo
9.
Cells ; 10(4)2021 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-33916770

RESUMO

Pregnancies carried by women with chronic hypertension are at increased risk of superimposed preeclampsia, but the placental pathways involved in disease progression remain poorly understood. In this study, we used the stroke-prone spontaneously hypertensive rat (SHRSP) model to investigate the placental mechanisms promoting superimposed preeclampsia, with focus on cellular stress and its influence on galectin-glycan circuits. Our analysis revealed that SHRSP placentas are characterized by a sustained activation of the cellular stress response, displaying significantly increased levels of markers of lipid peroxidation (i.e., thiobarbituric acid reactive substances (TBARS)) and protein nitration and defective antioxidant enzyme expression as early as gestation day 14 (which marks disease onset). Further, lectin profiling showed that such redox imbalance was associated with marked alterations of the placental glycocode, including a prominent decrease of core 1 O-glycan expression in trophoblasts and increased decidual levels of sialylation in SHRSP placentas. We also observed significant changes in the expression of galectins 1, 3 and 9 with pregnancy progression, highlighting the important role of the galectin signature as dynamic interpreters of placental microenvironmental challenges. Collectively, our findings uncover a new role for the glycoredox balance in the pathogenesis of superimposed preeclampsia representing a promising target for interventions in hypertensive disorders of pregnancy.


Assuntos
Progressão da Doença , Placenta/metabolismo , Polissacarídeos/metabolismo , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Feminino , Galectinas/metabolismo , Glicosilação , Modelos Biológicos , Oxirredução , Fenótipo , Gravidez , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Estresse Fisiológico , Fatores de Tempo
10.
Front Immunol ; 12: 784473, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975875

RESUMO

Lectin-glycan interactions, in particular those mediated by the galectin family, regulate many processes required for a successful pregnancy. Over the past decades, increasing evidence gathered from in vitro and in vivo experiments indicate that members of the galectin family specifically bind to both intracellular and membrane bound carbohydrate ligands regulating angiogenesis, immune-cell adaptations required to tolerate the fetal semi-allograft and mammalian embryogenesis. Therefore, galectins play important roles in fetal development and placentation contributing to maternal and fetal health. This review discusses the expression and role of galectins during the course of pregnancy, with an emphasis on maternal immune adaptions and galectin-glycan interactions uncovered in the recent years. In addition, we summarize the galectin fingerprints associated with pathological gestation with particular focus on preeclampsia.


Assuntos
Adaptação Fisiológica , Desenvolvimento Fetal/fisiologia , Galectinas/fisiologia , Placentação/fisiologia , Feminino , Galectinas/química , Glicoproteínas/fisiologia , Humanos , Gravidez
11.
Sci Rep ; 10(1): 15243, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32943699

RESUMO

Early postnatal life is characterized by a critical time period in which the developing neonatal immune system transitions from passive immunity, induced by protective maternal antibodies, to the competence of a fully functioning immune system. The inflammatory capability of both maternal and neonatal antibodies is governed by N-linked glycosylation of the Fc region, and though this has been examined extensively in adults, there is currently little information regarding antibody glycosylation patterns during early postnatal life. To characterize the murine IgG Fc glycosylation profile during early life, we used nano-LC-ESI-Qq-TOF mass spectrometry analysis to assess subclass specific Asn-297 glycosylation patterns in the serum of BALB/c mice from 5-60 days of age. From birth to adulthood, we observed a decline in proinflammatory Fc glycosylation in all IgG subclasses. This was shown by significantly reduced agalactosylated and monogalactosylated structures combined with increased sialylation after weaning at 45 and 60 days of age. This information indicates that the transition between neonatal life and adulthood in mice is accompanied by reduction of inflammatory IgG antibodies. Our study contributes to a growing body of literature indicating the importance of IgG Fc glycosylation and its association with inflammation during different life stages.


Assuntos
Sistema Imunitário/crescimento & desenvolvimento , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Glicosilação , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Fragmentos Fc das Imunoglobulinas/classificação , Imunoglobulina G/classificação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Espectrometria de Massas por Ionização por Electrospray
12.
Front Immunol ; 11: 1316, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32760395

RESUMO

Immune cells [e. g., dendritic cells (DC) and natural killer (NK) cells] are critical players during the pre-placentation stage for successful mammalian pregnancy. Proper placental and fetal development relies on balanced DC-NK cell interactions regulating immune cell homing, maternal vascular expansion, and trophoblast functions. Previously, we showed that in vivo disruption of the uterine NK cell-DC balance interferes with the decidualization process, with subsequent impact on placental and fetal development leading to fetal growth restriction. Glycans are essential determinants of reproductive health and the glycocode expressed in a particular compartment (e.g., placenta) is highly dependent on the cell type and its developmental and pathological state. Here, we aimed to investigate the maternal and placental glycovariation during the pre- and post-placentation period associated with disruption of the NK cell-DC dynamics during early pregnancy. We observed that depletion of NK cells was associated with significant increases of O- and N-linked glycosylation and sialylation in the decidual vascular zone during the pre-placental period, followed by downregulation of core 1 and poly-LacNAc extended O-glycans and increased expression of branched N-glycans affecting mainly the placental giant cells and spongiotrophoblasts of the junctional zone. On the other hand, expansion of DC induced a milder increase of Tn antigen (truncated form of mucin-type O-glycans) and branched N-glycan expression in the vascular zone, with only modest changes in the glycosylation pattern during the post-placentation period. In both groups, this spatiotemporal variation in the glycosylation pattern of the implantation site was accompanied by corresponding changes in galectin-1 expression. Our results show that pre- and post- placentation implantation sites have a differential glycopattern upon disruption of the NK cell-DC dynamics, suggesting that immune imbalance early in gestation impacts placentation and fetal development by directly influencing the placental glycocode.


Assuntos
Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Placenta/imunologia , Placentação , Animais , Feminino , Glicosilação , Masculino , Camundongos Endogâmicos BALB C , Gravidez
13.
Semin Immunopathol ; 42(4): 469-486, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32601855

RESUMO

Growing evidence suggests that galectins, an evolutionarily conserved family of glycan-binding proteins, fulfill key roles in pregnancy including blastocyst implantation, maternal-fetal immune tolerance, placental development, and maternal vascular expansion, thereby establishing a healthy environment for the growing fetus. In this review, we comprehensively present the function of galectins in shaping cellular circuits that characterize a healthy pregnancy. We describe the current understanding of galectins in term and preterm labor and discuss how the galectin-glycan circuits contribute to key immunological pathways sustaining maternal tolerance and preventing microbial infections. A deeper understanding of the glycoimmune pathways regulating early events in preterm birth could offer the broader translational potential for the treatment of this devastating syndrome.


Assuntos
Galectinas , Nascimento Prematuro , Implantação do Embrião , Feminino , Humanos , Tolerância Imunológica , Recém-Nascido , Polissacarídeos , Gravidez
14.
Cell Death Dis ; 11(7): 560, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32703931

RESUMO

Fetal growth restriction (FGR) is the most common pregnancy complication in developed countries. Pregnancies affected by FGR, frequently concur with complications and high risk of neonatal morbidity and mortality. To date, no approved treatment is available for pregnant women affected with FGR. The objective of this study was to investigate the contribution of galectin-3 (gal-3), a ß-galactoside binding protein involved in pregnancy, placental function and fetal growth. We demonstrated that lack of gal-3 during mouse pregnancy leads to placental dysfunction and drives FGR in the absence of a maternal preeclampsia syndrome. Analysis of gal-3 deficient dams revealed placental inflammation and malperfusion, as well as uterine natural killer cell infiltration with aberrant activation. Our results also show that FGR is associated with a failure to increase maternal circulating gal-3 levels during the second and third trimester in human pregnancies. Placentas from human pregnancies affected by FGR displayed lower gal-3 expression, which correlated with placental dysfunction. These data highlight the importance of gal-3 in the promotion of proper placental function, as its absence leads to placental disease and subsequent FGR.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Galectina 3/deficiência , Galectina 3/metabolismo , Insuficiência Placentária/metabolismo , Animais , Feminino , Desenvolvimento Fetal , Humanos , Masculino , Camundongos Endogâmicos C57BL , Placentação , Gravidez , Fatores de Risco
15.
Am J Reprod Immunol ; 84(6): e13311, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32691950

RESUMO

PROBLEM: From conception, a delicate regulation of galectins, a family of carbohydrate-binding proteins, is established to ensure maternal immune tolerance in pregnancy. Though galectin-3 (gal-3), the only chimera-type galectin, is abundantly expressed at the feto-maternal interface; the physiological role of this lectin during pregnancy remains to be fully elucidated and requires further investigation. METHOD OF STUDY: In this study, we analyzed serum gal-3 levels during the course of healthy gestation. Trophoblast functions were evaluated upon gal-3 exogenous stimulation using trophoblastic cell lines (e.g. , HIPEC65, SGHPL-4, and BeWo cells). Finally, we investigated variations in peripheral gal-3 levels associated with the development of spontaneous abortion and gestational diabetes mellitus (GDM). RESULTS: Gal-3 circulating levels increased as normal pregnancy progressed. In vitro experiments showed that exogenous gal-3 positively regulated trophoblast functions inducing invasion, tube formation, and fusion. Compared with normal pregnant women, circulating gal-3 levels were significantly decreased in patients who developed GDM. CONCLUSION: Our results reveal a physiological role for gal-3 during pregnancy, promoting proper trophoblast functions associated with healthy gestation. GDM is associated with a failure to increase circulating gal-3 levels late in gestation. Thus, dysregulation of gal-3 may indicate a contribution of the chimera-type lectin to this adverse pregnancy outcome.


Assuntos
Aborto Espontâneo/metabolismo , Diabetes Gestacional/metabolismo , Galectina 3/metabolismo , Gravidez/metabolismo , Trofoblastos/metabolismo , Linhagem Celular , Feminino , Galectina 3/genética , Humanos , Tolerância Imunológica , Circulação Placentária , Trofoblastos/patologia
16.
Front Immunol ; 11: 599104, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33717050

RESUMO

Galectin (gal)-1, -3, and -9 are members of a family of glycan binding proteins that mediate complex interactions between decidual, inflammatory and trophoblast cells modulating several processes during gestation, control of the maternal immune system, and parturition. Their immunomodulatory role in preterm birth and postnatal expression in preterm infants is unknown. We performed a single center prospective study of 170 preterm infants with a gestational age below 35 weeks. Peripheral venous blood samples were collected during the neonatal period and galectin-1, -3, and -9 were determined by ELISA. We noted a strong decline of circulating gal-1 and -3 levels but not gal-9 from birth to day 7 of life. There was an inverse correlation of gal-1 and -3 levels at birth with gestational age. Gal-1 levels were remarkably increased in infants born to amniotic infection syndrome (AIS), which was also observed for gal-9 levels. Infants who developed early-onset sepsis had higher levels of gal-3 at day 1 as compared to unaffected infants. Our observational data imply that galectin-1, -3, and -9 levels are elevated in preterm infants born in an inflammatory milieu such as AIS or EOS. Future studies need to address whether galectins mediate inflammation-induced preterm birth and could therefore be a target for clinical trials.


Assuntos
Âmnio , Galectinas/sangue , Recém-Nascido Prematuro/sangue , Infecções/sangue , Feminino , Humanos , Recém-Nascido , Masculino , Estudos Prospectivos
17.
Front Immunol ; 10: 1166, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231368

RESUMO

Galectins are a phylogenetically conserved family of soluble ß-galactoside binding proteins, consisting of 15 different types, each with a specific function. Galectins contribute to placentation by regulating trophoblast development, migration, and invasion during early pregnancy. In addition, galectins are critical players regulating maternal immune tolerance to the embedded embryo. Recently, the role of galectins in angiogenesis during decidualization and in placenta formation has gained attention. Altered expression of galectins is associated with abnormal pregnancies and infertility. This review focuses on the role of galectins in pregnancy-associated processes and discusses the relevance of galectin-glycan interactions as potential therapeutic targets in pregnancy disorders.


Assuntos
Galectinas/fisiologia , Polissacarídeos/metabolismo , Proteínas da Gravidez/fisiologia , Animais , Sequência de Carboidratos , Mapeamento Cromossômico , Dimerização , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Feminino , Galectinas/química , Galectinas/genética , Glicosilação , Humanos , Troca Materno-Fetal/fisiologia , Neovascularização Fisiológica/fisiologia , Placentação/fisiologia , Polissacarídeos/química , Pré-Eclâmpsia/metabolismo , Gravidez , Processamento de Proteína Pós-Traducional , Relação Estrutura-Atividade , Especificidade por Substrato , Trofoblastos/metabolismo
18.
Shock ; 52(2): 264-273, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30044324

RESUMO

Sepsis constitutes one of the major causes of death in ICUs. In sepsis induced by gram-negative, although lipopolysaccharide (LPS) initially induces an exacerbated secretion of proinflammatory cytokines leading to endotoxic shock and death resembling a septic shock, it is also capable of inducing refractoriness to subsequent challenge with LPS, a state known as endotoxin tolerance, which is considered the initial step of the immunosuppression found in septic patients. As we previously demonstrated the importance of glucocorticoids in endotoxin tolerance, the aim of this study was to evaluate the contribution of Interleukin-10 (IL-10) both in the endotoxic shock and in the development of the tolerance and its relationship with glucocorticoids. Our results show that, upon LPS challenge, IL-10 knockout mice (KO) mice had an enhanced LPS sensitivity, along with elevated levels of proinflammatory cytokines as tumor necrosis factor-α, IL-12 and interferon-γ, and enhanced tissue damage, despite the high levels of glucocorticoids. This effect may be because, in part, of the higher expression of tumor necrosis factor receptors in IL-10 KO mice. Further, the injection of dexamethasone did not protect IL-10 KO mice from a LPS lethal challenge. Although tolerance was achieved in the absence of IL-10, it was weaker and the elevated levels of glucocorticoids were not able to reverse the high sensitivity of IL-10 KO mice to LPS. Nevertheless, glucocorticoids would play a pivotal role in the establishment and maintenance of this partial tolerance in IL-10 KO mice. Finally, our results show that IL-10 and glucocorticoids could act in a bidirectional way influencing the inflammatory and anti-inflammatory periods.


Assuntos
Endotoxinas/toxicidade , Glucocorticoides/farmacologia , Interleucina-10/deficiência , Choque Séptico/tratamento farmacológico , Animais , Citocinas/metabolismo , Dexametasona/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Interleucina-10/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mifepristona/uso terapêutico , Choque Séptico/induzido quimicamente , Fator de Necrose Tumoral alfa/metabolismo
19.
Sci Rep ; 7(1): 2189, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28526846

RESUMO

DC-NK cell interactions are thought to influence the development of maternal tolerance and de novo angiogenesis during early gestation. However, it is unclear which mechanism ensures the cooperative dialogue between DC and NK cells at the feto-maternal interface. In this article, we show that uterine NK cells are the key source of IL-10 that is required to regulate DC phenotype and pregnancy success. Upon in vivo expansion of DC during early gestation, NK cells expressed increased levels of IL-10. Exogenous administration of IL-10 was sufficient to overcome early pregnancy failure in dams treated to achieve simultaneous DC expansion and NK cell depletion. Remarkably, DC expansion in IL-10-/- dams provoked pregnancy loss, which could be abrogated by the adoptive transfer of IL-10+/+ NK cells and not by IL-10-/- NK cells. Furthermore, the IL-10 expressing NK cells markedly enhanced angiogenic responses and placental development in DC expanded IL-10-/- dams. Thus, the capacity of NK cells to secrete IL-10 plays a unique role facilitating the DC-NK cell dialogue during the establishment of a healthy gestation.


Assuntos
Comunicação Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interleucina-10/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Troca Materno-Fetal , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Animais , Feminino , Desenvolvimento Fetal/genética , Humanos , Camundongos , Camundongos Knockout , Gravidez , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
20.
J Reprod Immunol ; 114: 38-43, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26956510

RESUMO

Galectin-1 (gal-1), a member of a family of conserved ß-galactoside-binding proteins, has been shown to exert a key role during gestation. Though gal-1 is expressed at higher levels in the placenta from HELLP patients, it is still poorly understood whether systemic gal-1 levels also differ in HELLP patients. In the present study, we evaluated the systemic expression of gal-1, together with the angiogenic factors, placental growth factor (PlGF) and soluble fms-like tyrosine kinase 1 (sFlt-1) in conjunction with HELLP syndrome severity. Systemic levels of gal-1 and sFlt-1 were elevated in patients with both early- and late-onset HELLP syndrome as compared to healthy controls. In contrast, peripheral PlGF levels were decreased in early- and late-onset HELLP. A positive correlation between systemic gal-1 levels and sFlt-1/PlGF ratios was found in early onset HELLP patients. Our results show that HELLP syndrome is associated with increased circulating levels of gal-1; integrating systemic gal-1 measurements into the diagnostic analyses of pregnant women may provide more effective prediction of HELLP syndrome development.


Assuntos
Galectina 1/sangue , Síndrome HELLP/sangue , Segundo Trimestre da Gravidez/sangue , Terceiro Trimestre da Gravidez/sangue , Adulto , Feminino , Galectina 1/imunologia , Síndrome HELLP/imunologia , Humanos , Gravidez , Segundo Trimestre da Gravidez/imunologia , Terceiro Trimestre da Gravidez/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA