Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 8(37): 61001-61010, 2017 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-28977841

RESUMO

Endothelial injury and dysfunction followed by endothelial activation and inflammatory cell recruitment are factors contributing to the initiation and progression of atherosclerosis. Oxidized low-density lipoprotein (ox-LDL) promotes inflammation during atherogenesis and lipid deposition in the arterial wall. We observed that stimulation of human umbilical vein endothelial cells (HUVECs) with ox-LDL activated pro-inflammatory cytokine production and apoptosis, inhibited cell migration, and upregulated T-cell immunoglobulin and mucin domain 3 (Tim-3) expression. Tim-3, in turn, protected HUVECs from ox-LDL-induced apoptosis via the JNK pathway and reversed the inhibition of migration. Tim-3 also inhibited ox-LDL-induced inflammatory cytokine production by suppressing NF-κB activation. In addition, Tim-3 increased production of type 2 T helper cells (Th2) and regulatory T cell (Treg)-associated cytokines. Blocking Tim-3 reversed its effects on the inflammatory response to ox-LDL. Thus, Tim-3 signaling may be a "self-control" mechanism in ox-LDL-triggered inflammation in HUVECs. These results identify Tim-3 as a factor in HUVEC activity and suggest its potential in the treatment of atherosclerosis.

2.
J Reprod Dev ; 63(3): 289-294, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28331165

RESUMO

During pregnancy, CD8+ T cells are important regulators in the balance of fetal tolerance and antiviral immunity. T-cell immunoglobulin mucin-3 (Tim-3) and programmed cell death-1 (PD-1) are well-recognized negative co-stimulatory molecules involved in viral persistence and tumor metastasis. Here, we demonstrate that CD8+ T cells co-expressing Tim-3 and PD-1 were down-regulated in the deciduae of female mice in abortion-prone matings compared with normal pregnant mice. In addition to their reduced numbers, the Tim-3+PD-1+CD8+ T cells produced lower levels of the anti-inflammatory cytokines interleukin (IL)-4 and IL-10, as well as a higher level of the pro-inflammatory cytokine interferon (IFN)-γ, relative to those from normal pregnancy. Furthermore, normal pregnant CBA/J females challenged with Tim-3- and/or PD-1-blocking antibodies were more susceptible to fetal resorption. These findings indicate that Tim-3 and PD-1 pathways play critical roles in regulating CD8+ T cell function and maintaining normal pregnancy.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Perda do Embrião , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Prenhez/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Animais , Feminino , Tolerância Imunológica , Interferon gama/metabolismo , Masculino , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Gravidez
3.
Trends Mol Med ; 23(1): 46-58, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27914866

RESUMO

Physiologically, a successful pregnancy requires the maternal immune system to recognize and tolerate the semiallogeneic fetus, and allow for normal invasion of trophoblasts. Thus, pregnancy complications are considered to be associated with dysfunctional maternal-fetal crosstalk. Co-signaling molecules are a group of cell surface molecules that positively or negatively modulate the immune response. Well studied in the fields of oncology and transplantation, they are also suggested to be involved in maternal-fetal crosstalk. Here, we review the latest knowledge on the expression and function of such co-signaling molecules, highlighting their immunoregulatory roles in maternal-fetal tolerance and decidual vascular remodeling, and their involvement in pathological pregnancies. This review may instruct future basic research on, and clinical applications for, maternal-fetal immunity.


Assuntos
Feto/imunologia , Troca Materno-Fetal , Complicações na Gravidez/imunologia , Gravidez/imunologia , Animais , Decídua/irrigação sanguínea , Decídua/imunologia , Feminino , Humanos , Tolerância Imunológica , Imunidade , Camundongos , Transdução de Sinais , Trofoblastos/imunologia
4.
Cell Mol Immunol ; 13(1): 73-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25578313

RESUMO

Decidual natural killer (dNK) cells actively participate in the establishment and maintenance of maternal-fetal immune tolerance and act as local guardians against infection. However, how dNK cells maintain the immune balance between tolerance and anti-infection immune responses during pregnancy remains unknown. Here, we demonstrated that the inhibitory molecule T-cell immunoglobulin domain and mucin domain-containing molecule-3 (Tim-3) are expressed on over 60% of dNK cells. Tim-3(+) dNK cells display higher interleukin (IL)-4 and lower tumor necrosis factor (TNF)-α and perforin production. Human trophoblast cells can induce the transformation of peripheral NK cells into a dNK-like phenotype via the secretion of galectin-9 (Gal-9) and the interaction between Gal-9 and Tim-3. In addition, trophoblasts inhibit lipopolysaccharide (LPS)-induced pro-inflammatory cytokine and perforin production by dNK cells, which can be attenuated by Tim-3 neutralizing antibodies. Interestingly, a decreased percentage of Tim-3-expressing dNK cells were observed in human miscarriages and murine abortion-prone models. Moreover, T helper (Th)2-type cytokines were decreased and Th1-type cytokines were increased in Tim-3(+) but not Tim-3(-) dNK cells from human and mouse miscarriages. Therefore, our results suggest that the Gal-9/Tim-3 signal is important for the regulation of dNK cell function, which is beneficial for the maintenance of a normal pregnancy.


Assuntos
Aborto Espontâneo/genética , Galectinas/imunologia , Células Matadoras Naturais/imunologia , Proteínas de Membrana/imunologia , Trofoblastos/imunologia , Aborto Espontâneo/imunologia , Aborto Espontâneo/patologia , Adulto , Animais , Anticorpos Neutralizantes/farmacologia , Técnicas de Cocultura , Decídua/imunologia , Decídua/patologia , Feminino , Galectinas/genética , Regulação da Expressão Gênica , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Tolerância Imunológica , Interleucina-4/genética , Interleucina-4/imunologia , Células Matadoras Naturais/patologia , Lipopolissacarídeos/farmacologia , Troca Materno-Fetal/imunologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Perforina/genética , Perforina/imunologia , Gravidez , Cultura Primária de Células , Transdução de Sinais , Trofoblastos/patologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
5.
PLoS One ; 10(6): e0128523, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26035207

RESUMO

T cell-mediated immunity plays a significant role in the development of atherosclerosis (AS). There is increasing evidence that CD8+ T cells are also involved in AS but their exact roles remain unclear. The inhibitory receptors programmed cell death-1 (PD-1) and T cell immunoglobulin and mucin domain 3 (Tim-3) are well known inhibitory molecules that play a crucial role in regulating CD8+ T cell activation or tolerance. Here, we demonstrate that the co-expression of PD-1 and Tim-3 on CD8+ T cells is up-regulated in AS patients. PD-1+ Tim-3+ CD8+ T cells are enriched for within the central T (TCM) cell subset, with high proliferative activity and CD127 expression. Co-expression of PD-1 and Tim-3 on CD8+ T cells is associated with increased anti-atherogenic cytokine production as well as decreased pro-atherogenic cytokine production. Blockade of PD-1 and Tim-3 results in a decrease of anti-atherogenic cytokine production by PD-1+ Tim-3+ CD8+ T cells and in an augmentation of TNF-α and IFN-γ production. These findings highlight the important role of the PD-1 and Tim-3 pathways in regulating CD8+ T cells function in human AS.


Assuntos
Aterosclerose/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/imunologia , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Proteínas de Membrana/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Tolerância Imunológica , Técnicas Imunoenzimáticas , Linfócitos/metabolismo , Linfócitos/patologia , Masculino , Camundongos , Pessoa de Meia-Idade
6.
Cell Mol Immunol ; 12(1): 77-86, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24793405

RESUMO

Decidual natural killer (dNK) cells are believed to be critical for maintaining maternal/fetal tolerance and regulating placental vascular remodeling based upon their abundance and unique phenotype during early pregnancy. However, the mechanism for how the dNK cells play such important roles in successful pregnancy remains undefined. Here, we identified a subtype of dNK cells characterized as having a CD3(-)CD56(bright)CD25(+) phenotype. We found that CD56(bright)CD25(+) NK cells preferentially localize to the maternal/fetal interface during early human pregnancy. CD25(+) dNK cells account for approximately 75% of CD25-expressing decidual immune cells (DICs). However, less than 5% of CD25-positive peripheral blood mononuclear cells are CD25(+) NK cells. Furthermore, CD25(+) and CD25(-) dNK cells exhibit distinct phenotypes: CD25(+) dNK cells display a more activated phenotype and greater cytokine-secreting capacity. Interestingly, coculture of peripheral NK (pNK) cells with primary trophoblasts upregulates the percentage of CD25-expressing pNK cells, resulting in increased expression of activation markers and cytokine production by pNK cells. In addition, we demonstrated that the CXCL12/CXCR4 axis is crucial for the recruitment of CD25(+) dNK cells and contributes to the accumulation of CD3(-)CD56(bright)CD25(+) dNK cells at the maternal/fetal interface. Thus, our data reveal that the crosstalk between trophoblasts and pNK cells leads to the accumulation of CD3(-)CD56(bright)CD25(+) dNK cells, which exert a regulating effect at the maternal/fetal interface.


Assuntos
Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Trofoblastos/imunologia , Antígeno CD56/metabolismo , Comunicação Celular , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Citocinas/metabolismo , Feminino , Humanos , Imunofenotipagem , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Ativação Linfocitária , Troca Materno-Fetal/imunologia , Gravidez/imunologia , Receptores CXCR4
7.
Cell Mol Immunol ; 11(5): 438-48, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25109684

RESUMO

Embryos express paternal antigens that are foreign to the mother, but the mother provides a special immune milieu at the fetal-maternal interface to permit rather than reject the embryo growth in the uterus until parturition by establishing precise crosstalk between the mother and the fetus. There are unanswered questions in the maintenance of pregnancy, including the poorly understood phenomenon of maternal tolerance to the allogeneic conceptus, and the remarkable biological roles of placental trophoblasts that invade the uterine wall. Chemokines are multifunctional molecules initially described as having a role in leukocyte trafficking and later found to participate in developmental processes such as differentiation and directed migration. It is increasingly evident that the gestational uterine microenvironment is characterized, at least in part, by the differential expression and secretion of chemokines that induce selective trafficking of leukocyte subsets to the maternal-fetal interface and regulate multiple events that are closely associated with normal pregnancy. Here, we review the expression and function of chemokines and their receptors at the maternal-fetal interface, with a special focus on chemokine as a key component in trophoblast invasiveness and placental angiogenesis, recruitment and instruction of immune cells so as to form a fetus-supporting milieu during pregnancy. The chemokine network is also involved in pregnancy complications.


Assuntos
Quimiocinas/imunologia , Troca Materno-Fetal/imunologia , Complicações na Gravidez/imunologia , Trofoblastos/imunologia , Movimento Celular , Feminino , Humanos , Tolerância Imunológica , Isoantígenos/imunologia , Neovascularização Fisiológica , Gravidez
8.
J Immunol ; 192(4): 1502-11, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24453244

RESUMO

Physiological pregnancy requires the maternal immune system to recognize and tolerate embryonic Ags. Although multiple mechanisms have been proposed, it is not yet clear how the fetus evades the maternal immune system. In this article, we demonstrate that trophoblast-derived thymic stromal lymphopoietin (TSLP) instructs decidual CD11c(+) dendritic cells (dDCs)with increased costimulatory molecules; MHC class II; and Th2/3-type, but not Th1-type, cytokines. TSLP-activated dDCs induce proliferation and differentiation of decidual CD4(+)CD25(-) T cells into CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) through TGF-ß1. TSLP-activated dDC-induced Tregs display immunosuppressive features and express Th2-type cytokines. In addition, decidual CD4(+)CD25(+)FOXP3(+) Tregs promote invasiveness and HLA-G expression of trophoblasts, resulting in preferential production of Th2 cytokines and reduced cytotoxicity in decidual CD56(bright)CD16(-) NK cells. Of interest, decreased TSLP expression and reduced numbers of Tregs were observed at the maternal-fetal interface during miscarriage. Our study identifies a novel feedback loop between embryo-derived trophoblasts and maternal decidual leukocytes, which induces a tolerogenic immune response to ensure a successful pregnancy.


Assuntos
Citocinas/metabolismo , Células Dendríticas/metabolismo , Histocompatibilidade Materno-Fetal/imunologia , Linfócitos T Reguladores/metabolismo , Aborto Espontâneo/metabolismo , Adulto , Antígeno CD11c/imunologia , Antígenos CD4/metabolismo , Antígeno CD56/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Citocinas/imunologia , Decídua/citologia , Decídua/metabolismo , Células Dendríticas/imunologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Antígenos de Histocompatibilidade Classe II , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Células Matadoras Naturais/imunologia , Gravidez , Receptores de IgG/metabolismo , Células Th2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Trofoblastos/imunologia , Trofoblastos/metabolismo , Adulto Jovem , Linfopoietina do Estroma do Timo
9.
Int J Clin Exp Pathol ; 6(10): 1999-2010, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24133577

RESUMO

Our previous studies have demonstrated that cyclosporin A (CsA) promotes the proliferation and migration of human trophoblasts via the mitgen-activated protein kinase-3/1 (MAPK3/1) pathway. In the present study, we further investigated the role of nuclear factor (NF)-κB in the CsA-induced trophoblast proliferating cell nuclear antigen (PCNA) expression and migration, and its relationship to MAPK3/1 signal. Flow cytometry was used to analyze the expression of PCNA in trophoblasts. The migration of human primary trophoblasts was determined by wound-healing assay and transwell migration assay. Western blot analysis was performed to evaluate the activation of NF-κB p65 and NF-κB inhibitory protein I-κB in human trophoblasts. We found that treatment with CsA promotes PCNA expression and migration of human trophoblast in a dose-associated manner. Blocking of the MAPK3/1 signal abrogated the enhanced PCNA expression and migration in trophoblasts by CsA. In addition, CsA increased the phosphorylation of NF-κB p65 and the inhibitor I-κB in human trophoblasts in a time-related manner. Pretreatment with MAPK3/1 inhibitor U0126 abrogated the phosphorylation of NF-κB p65 and I-κB. Accordingly, the CsA-induced enhancement of PCNA expression and migration in trophoblasts was also decreased. This CsA-induced enhancement in the expression and migration of trophoblasts was abolished by pretreatment with pyrrolidine dithiocarbamate, a specific NF-κB inhibitor. Thus, our results suggest that CsA promotes PCNA expression and migration of human trophoblasts via MAPK-mediated NF-κB activation.


Assuntos
Movimento Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Trofoblastos/citologia , Butadienos/farmacologia , Movimento Celular/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Placenta/citologia , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
10.
PLoS One ; 8(9): e74812, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069351

RESUMO

Hyaluronan (HA) and its receptor CD44 are expressed at the maternal-fetal interface, but its role in early pregnancy remains unclear. Here, we found that primary decidual stromal cells (DSCs) continuously secreted HA and expressed its receptor CD44. Pregnancy-associated hormones up-regulated HA synthetase (HAS) 2 transcription and HA release from DSCs. High molecular weight-HA (HMW-HA), but not medium molecular weight (MMW-HA) or low molecular weight (LMW-HA), promoted proliferation and inhibited apoptosis of DSCs in a CD44-dependent manner. The in-cell Western analysis revealed HMW-HA activated PI3K/AKT and mitogen-activated protein kinase (MAPK)/ERK1/2 signaling pathways time-dependently. Blocking these pathways by specific inhibitor LY294002 or U0126 abrogated HMW-HA-regulated DSc proliferation and apoptosis. Finally, we have found that HA content, HA molecular weight, HAS2 mRNA level, and CD44 expression were significantly decreased in DSCs from unexplained miscarriage compared with the normal pregnancy. Collectively, our results indicate that higher level and greater molecular mass of HA at maternal-fetal interface contributes to DSc growth and maintenance of DSCs in human early pregnancy.


Assuntos
Decídua/citologia , Decídua/metabolismo , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Células Estromais/metabolismo , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Adulto , Apoptose , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hormônios Esteroides Gonadais/farmacologia , Humanos , Hialuronan Sintases , Ácido Hialurônico/química , Imuno-Histoquímica , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peso Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Transcrição Gênica , Adulto Jovem
11.
Mol Hum Reprod ; 19(10): 676-86, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23737337

RESUMO

Spontaneous abortion is the most common complication of pregnancy. Immune activation and the subsequent inflammation-induced tissue injury are often observed at the maternal-fetal interface as the final pathological assault in recurrent spontaneous abortion. However, the precise mechanisms responsible for spontaneous abortion involving inflammation are not fully understood. Chemokine CCL28 and its receptors CCR3 and CCR10 are important regulators in inflammatory process. Here, we examined the expression of CCL28 and its receptors in decidual stromal cells (DSCs) by immunochemistry and flow cytometry (FCM), and compared their expression level in DSCs from normal pregnancy versus spontaneous abortion, and their relationship to inflammatory cytokines production by DSCs. We further analyzed regulation of the pro-inflammatory cytokines on CCL28 expression in DSCs by real-time polymerase chain reaction, In-cell Western and FCM. The effects of CCL28-CCR3/CCR10 interaction on DSC apoptosis was investigated by Annexin V staining and FCM analysis or DAPI staining and nuclear morphology. Higher levels of the inflammatory cytokines interleukin (IL)-1ß, IL-17A and tumor necrosis factor-α, and increased CCR3/CCR10 expression were observed in DSCs from spontaneous abortion compared with normal pregnancy. Treatment with inflammatory cytokines differently affected CCL28 and CCR3/CCR10 expression in DSCs. Human recombinant CCL28 promoted DSC apoptosis, which was eliminated by pretreatment with neutralizing antibodies against CCR3/CCR10 and CCL28. However, CCL28 did not affect DSC growth. These results suggest that the inflammation-promoted up-regulation of CCL28 and its receptors interaction in DSCs is involved in human spontaneous abortion via inducing DSC apoptosis.


Assuntos
Aborto Espontâneo/metabolismo , Quimiocinas CC/metabolismo , Decídua/citologia , Receptores CCR10/metabolismo , Receptores CCR3/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Aborto Espontâneo/genética , Adulto , Apoptose/efeitos dos fármacos , Apoptose/genética , Células Cultivadas , Quimiocinas CC/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas In Vitro , Interleucina-17/farmacologia , Interleucina-1beta/farmacologia , Gravidez , Receptores CCR10/genética , Receptores CCR3/genética , Células Estromais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Adulto Jovem
12.
PLoS One ; 7(9): e45275, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028901

RESUMO

Our previous study has demonstrated that cyclosporine A (CsA) administration in vivo induces Th2 bias at the maternal-fetal interface, leading to improved murine pregnancy outcomes. Here, we investigated how CsA treatment in vitro induced Th2 bias at the human maternal-fetal interface in early pregnancy. The cell co-culture in vitro in different combination of component cells at the maternal-fetal interface was established to investigate the regulation of CsA on cytokine production from the interaction of these cells. It was found that interferon (IFN)-γ was produced only by decidual immune cells (DICs), and not by trophoblasts or decidual stromal cells (DSCs); all these cells secreted interleukin (IL)-4, IL-10, and tumor necrosis factor (TNF)-α. Treatment with CsA completely blocked IFN-γ production in DICs and inhibited TNF-α production in all examined cells. CsA increased IL-10 and IL-4 production in trophoblasts co-cultured with DSCs and DICs although CsA treatment did not affect IL-10 or IL-4 production in any of the cells when cultured alone. These results suggest that CsA promotes Th2 bias at the maternal-fetal interface by increasing Th2-type cytokine production in trophoblasts with the aid of DSCs and DICs, while inhibiting Th1-type cytokine production in DICs and TNF-α production in all investigated cells. Our study might be useful in clinical therapeutics for spontaneous pregnancy wastage and other pregnancy complications.


Assuntos
Ciclosporina/farmacologia , Decídua/metabolismo , Imunossupressores/farmacologia , Células Th1/metabolismo , Células Th2/metabolismo , Trofoblastos/metabolismo , Adulto , Comunicação Celular , Técnicas de Cocultura , Decídua/citologia , Decídua/efeitos dos fármacos , Feminino , Humanos , Interferon gama/biossíntese , Interleucina-10/biossíntese , Interleucina-4/biossíntese , Gravidez , Primeiro Trimestre da Gravidez , Células Th1/citologia , Células Th1/efeitos dos fármacos , Equilíbrio Th1-Th2 , Células Th2/citologia , Células Th2/efeitos dos fármacos , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese
13.
Cell Mol Immunol ; 9(5): 423-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22885527

RESUMO

The regulatory mechanism of Th2 bias at the maternal/fetal interface remains unclear. In this study, we characterized cytokine production in decidual stromal cells (DSCs), decidual immune cells (DICs) and embryo-derived trophoblast cells, and investigated the regulation of CXCL12/CXCR4 interaction on Th2 bias at the maternal/fetal interface in early human pregnancy. We found differential production of Th1-type and Th2-type cytokines by trophoblasts, DSCs and DICs. The secretion of these cytokines varied in different cell cocultures, conduced to Th2 bias. Flow cytometry showed that coculture of trophoblasts with DSCs and DICs significantly increased IL-4 and IL-10 production in trophoblasts, and IL-10 production in DSCs. However, the coculture of trophoblasts with DSCs and DICs significantly increased interferon (IFN)-γ expression in DSCs, and tumor-necrosis factor (TNF)-α expression in DICs. No change was seen in Th1-type cytokine production in trophoblasts, and in Th2-type cytokine production in DICs in all cocultures. Furthermore, pre-treatment with anti-CXCR4 neutralizing antibody upregulated the production of the Th1-type cytokines IFN-γ and TNF-α, and downregulated the production of the Th2-type cytokines IL-4 and IL-10, in trophoblasts, DSCs, DICs or their cocultures. Interestingly, rhCXCL12 inhibited production of the Th1-type cytokine TNF-α and enhanced the expression of the Th2-type cytokines such as IL-4 and IL-10 in DICs; this effect was abrogated by anti-CXCR4 antibody. Our present study has elucidated the individual contributions of component cells to the shaping of Th2 bias, and uncovered a complicated cross-talk via the CXCL12/CXCR4 signal at the maternal/fetal interface in early human pregnancy.


Assuntos
Quimiocina CXCL12/metabolismo , Decídua/metabolismo , Gravidez/metabolismo , Receptores CXCR4/metabolismo , Trofoblastos/metabolismo , Adulto , Células Cultivadas , Técnicas de Cocultura , Citocinas/biossíntese , Decídua/citologia , Decídua/imunologia , Feminino , Citometria de Fluxo , Humanos , Gravidez/imunologia , Primeiro Trimestre da Gravidez , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Equilíbrio Th1-Th2 , Trofoblastos/citologia , Trofoblastos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA