Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Reprod Biomed Online ; 44(5): 803-816, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35339367

RESUMEN

RESEARCH QUESTION: Increased granulosa cell division is associated with abnormal folliculogenesis in polycystic ovary syndrome (PCOS). Lethal-7i microRNA (let-7i) may play an important role in the follicular development and granulosa cell growth; therefore is let-7i involved in PCOS pathogenesis? DESIGN: The expression of let-7i was measured in granulosa-luteal cells (GLC) from women with or without PCOS. A human granulosa cell line, KGN, was used for the functional study. Mimics and inhibitors of let-7i, lentiviruses expressing insulin-like growth factor 2 mRNA binding protein (IMP2), and small-interfering RNAs were transfected into KGN cells. KGN cell proliferation was determined by 5-ethynyl-2'-deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assays. The cell cycle and apoptosis were assessed by propidium iodide-annexin V (PI-A) staining and fluorescence-activated cell sorting. Oestradiol concentration was determined by enzyme-linked immunoassay. Bioinformatics analysis and luciferase reporter assay were applied to confirm the let-7i target genes. RESULTS: The study showed that let-7i was down-regulated in PCOS GLC (P = 0.001). Mimics of let-7i inhibited KGN proliferation (P = 0.001), and decreased aromatase expression (P = 0.030) and oestradiol production (P = 0.029), whereas let-7i inhibitors had the opposite effect. Bioinformatics analysis and quantitative real-time (qRT) PCR identified IMP2 as a target of let-7i (P = 0.021). qRT-PCR and western blot analysis indicated that IMP2 was up-regulated in GLC in women with PCOS (P = 0.001 and P = 0.044), and IMP2 expression was suppressed by let-7i in KGN cells (P < 0.001). Luciferase reporter assay results (P = 0.002), combined with the rescue assay, confirmed that let-7i inhibited KGN cell proliferation and reduced oestradiol concentration by directly targeting IMP2. CONCLUSIONS: let-7i was down-regulated in PCOS GLC. Overexpression of let-7i inhibited KGN cell proliferation and decreased oestradiol production in an IMP2-dependent manner, providing a new molecular mechanism for PCOS.


Asunto(s)
Células Lúteas , MicroARNs , Síndrome del Ovario Poliquístico , Femenino , Humanos , Apoptosis/fisiología , Proliferación Celular/fisiología , Estradiol/metabolismo , Células de la Granulosa/metabolismo , Células Lúteas/metabolismo , Células Lúteas/patología , MicroARNs/genética , MicroARNs/metabolismo , Síndrome del Ovario Poliquístico/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(3): 890-899, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593567

RESUMEN

The AML1-ETO fusion protein, generated by the t(8;21) chromosomal translocation, is causally involved in nearly 20% of acute myeloid leukemia (AML) cases. In leukemic cells, AML1-ETO resides in and functions through a stable protein complex, AML1-ETO-containing transcription factor complex (AETFC), that contains multiple transcription (co)factors. Among these AETFC components, HEB and E2A, two members of the ubiquitously expressed E proteins, directly interact with AML1-ETO, confer new DNA-binding capacity to AETFC, and are essential for leukemogenesis. However, the third E protein, E2-2, is specifically silenced in AML1-ETO-expressing leukemic cells, suggesting E2-2 as a negative factor of leukemogenesis. Indeed, ectopic expression of E2-2 selectively inhibits the growth of AML1-ETO-expressing leukemic cells, and this inhibition requires the bHLH DNA-binding domain. RNA-seq and ChIP-seq analyses reveal that, despite some overlap, the three E proteins differentially regulate many target genes. In particular, studies show that E2-2 both redistributes AETFC to, and activates, some genes associated with dendritic cell differentiation and represses MYC target genes. In AML patients, the expression of E2-2 is relatively lower in the t(8;21) subtype, and an E2-2 target gene, THPO, is identified as a potential predictor of relapse. In a mouse model of human t(8;21) leukemia, E2-2 suppression accelerates leukemogenesis. Taken together, these results reveal that, in contrast to HEB and E2A, which facilitate AML1-ETO-mediated leukemogenesis, E2-2 compromises the function of AETFC and negatively regulates leukemogenesis. The three E proteins thus define a heterogeneity of AETFC, which improves our understanding of the precise mechanism of leukemogenesis and assists development of diagnostic/therapeutic strategies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Recurrencia
3.
Sheng Li Xue Bao ; 72(1): 11-19, 2020 Feb 25.
Artículo en Zh | MEDLINE | ID: mdl-32099980

RESUMEN

Immune tolerance at maternal-fetal interface is the basis for establishment and maintenance of successful pregnancy. T cells are pivotal compositions of uterine decidual immune cells, which are required to mediate anti-infection immunity and protect embryos from external antigens attack. T cells also participate in the complex immune regulation process of maternal acceptance of semi-allogeneic embryos, and play an important role in regulating embryo implantation and maintaining pregnancy. Its dysfunction may lead to early pregnancy failures or mid-late pregnancy complications. This review summarizes the compositions, phenotypic characteristics and functions of decidual T cells at the maternal-fetal interface in recent years, and further describes the regulation of decidual CD4+ and CD8+ T cells in maternal-fetal immune tolerance as well as the molecular mechanisms of abnormal regulation leading to early pregnancy failures. Through the in-depth understanding the mechanism of maternal-fetal immune regulation, it supplies a novel concept on maternal-fetal immune tolerance and new clues for the immunotherapy of pregnancy-related diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Decidua/inmunología , Tolerancia Inmunológica , Intercambio Materno-Fetal/inmunología , Femenino , Feto , Humanos , Embarazo
4.
Sheng Li Xue Bao ; 72(2): 227-234, 2020 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-32328616

RESUMEN

Adrenergic receptor (AR), one of the key receptors for nervous system, plays an important role in the immune microenvironment and the progression of many diseases. In recent years, the regulation of ARs and its signal on macrophages has become a research hotspot. Researchers found that ARs could exert different regulatory functions on macrophages in different microenvironments, which in turn affects occurrence and development of diseases such as tumor, heart failure, obesity, acute injury, infection and pregnancy-related diseases. This review summarizes the expression and functional regulation of ARs on macrophages, and the role of ARs in microenvironment of related diseases, which might provide new ideas for the treatments.


Asunto(s)
Enfermedad , Macrófagos/fisiología , Receptores Adrenérgicos/fisiología , Transducción de Señal , Humanos
5.
Immunol Cell Biol ; 95(8): 695-704, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28653669

RESUMEN

Decidual NK (dNK) cells, identified as CD56brightCD16-CD3-, account for ~70% of lymphocytes within the uterine wall during early pregnancy. Accumulating evidence suggests that tight interactions between placental trophoblasts and dNK cells are critical for trophoblast cell differentiation. However, the underlying mechanism remains to be explored in detail. In the present study, conditioned medium (CM) was collected from cultured primary human dNK cells. Primary cytotrophoblasts (CTBs) or the human trophoblast cell line HTR8/SVneo was treated with dNK-CM and co-cultured with human umbilical vein endothelial cells (HUVECs) in a three-dimensional Matrigel scaffold, and the formation of tube structures was dynamically monitored with live cell imaging. Trophoblast invasion was analyzed with a transwell invasion assay. The data demonstrated that the treatment of HTR8/SVneo cells or CTBs with dNK-CM remarkably promoted trophoblast invasion and tube formation in the presence of HUVECs. The epithelial marker E-cadherin was reduced, while the expression of endothelial markers NCAM, VE-cadherin and integrin ß1 was significantly promoted in the HTR8/SVneo cells upon treatment with dNK-CM. Antibody blocking experiments revealed that the dNK cells promoted trophoblast invasion through the production of IL-8 and HGF, and they induced trophoblast differentiation toward endothelial phenotype by producing VEGF-C and HGF. These results provide new evidence to clarify the finely tuned interactions between trophoblasts and dNK cells at the maternal-fetal interface.


Asunto(s)
Decidua/inmunología , Células Endoteliales/inmunología , Factor de Crecimiento de Hepatocito/metabolismo , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Antígeno CD56/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-8/metabolismo , Morfogénesis , Embarazo , Cultivo Primario de Células
6.
J Reprod Dev ; 63(3): 289-294, 2017 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-28331165

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Pérdida del Embrión , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Preñez/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Femenino , Tolerancia Inmunológica , Interferón gamma/metabolismo , Masculino , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Embarazo
7.
Exp Mol Pathol ; 100(3): 506-13, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27163202

RESUMEN

The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is frequently altered in human malignancies and Akt over-expression and/or activation induces malignant transformation and chemoresistance. However, the role of Akt in the mechanisms of chemoresistance remains elusive. Here we reported that cisplatin treatment of chemosensitive, but not resistant, ovarian cancer cells (OVCAs) markedly increased the cell proportion in sub-G1 phase. Cisplatin however caused a significant accumulation of the resistant cells in S and G2/M phases, which was associated with a rapid and sustained checkpoint kinase 1 (Chk1) activation. In contrast, while cisplatin also elicited a rapid activation of Chk1 in sensitive cells, it markedly decreased total ChK1 and phospho-Chk1 contents over 12 h. Over-expression of dominant negative (DN)-AKT alone increased phospho-Chk1 content, and induced G2/M arrest and apoptosis. However, it inhibited Chk1 activation and G2/M arrest with combination of cisplatin treatment, resulting in p53-independent apoptosis. Furthermore, the responses of the chemoresistant cells to cisplatin were attenuated with forced expression of constitutive active AKT2. Chk1 knock-down also facilitated cisplatin-induced apoptosis in chemoresistant cells. Our studies implicate that, in addition to its cell survival and anti-apoptotic actions, Akt might also play an important role in the regulation of G2-M transition, possibly via up-regulation of Chk1 activity and stability. These data provide strong support for the concept that Akt is important in cell cycle regulation in the control of chemosensitivity in OVCAs and offers an alternate regulatory pathway for the development of rationale therapy for cisplatin-resistant ovarian cancer.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN
8.
J Cardiovasc Pharmacol ; 68(4): 322-326, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760067

RESUMEN

Microvascular complications are the leading causes of acquired blindness, end-stage renal failure, and varieties of neuropathy associated with diabetes. Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, is involved in endothelial dysfunction, oxidative stress, and inflammation associated with the progression of diabetic microvascular complications. Elevated ADMA has been detected in experimental animals and patients with diabetic microangiopathy like retinopathy, nephropathy, and neuropathy. In the review, we focus on the role of ADMA in the pathobiology of major microvascular complications of diabetes.


Asunto(s)
Arginina/análogos & derivados , Diabetes Mellitus/metabolismo , Angiopatías Diabéticas/metabolismo , Animales , Arginina/metabolismo , Biomarcadores/metabolismo , Diabetes Mellitus/patología , Angiopatías Diabéticas/patología , Inhibidores Enzimáticos/metabolismo , Humanos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo
9.
J Immunol ; 192(4): 1502-11, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24453244

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/metabolismo , Histocompatibilidad Materno-Fetal/inmunología , Linfocitos T Reguladores/metabolismo , Aborto Espontáneo/metabolismo , Adulto , Antígeno CD11c/inmunología , Antígenos CD4/metabolismo , Antígeno CD56/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citocinas/inmunología , Decidua/citología , Decidua/metabolismo , Células Dendríticas/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Antígenos de Histocompatibilidad Clase II , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Embarazo , Receptores de IgG/metabolismo , Células Th2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Trofoblastos/inmunología , Trofoblastos/metabolismo , Adulto Joven , Linfopoyetina del Estroma Tímico
10.
Gynecol Endocrinol ; 30(12): 885-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25170777

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common endocrine disorder in women of reproductive age with anti-müllerian hormone (AMH) two to three times higher, but the mechanism of increased AMH, excessive follicles and follicle stagnation in PCOS still needs further research. METHODS: Female Sprague-Dawley rats were treated with a gavage of 1.0 mg/kg of letrozole carboxymethylcellulose solution once daily for 21 consecutive days. Serum steroid concentrations, ovarian morphology, ovarian expression of AMH and AMH-RII protein were determined and their relationships were studied. RESULTS: According to the morphology and endocrinology, the letrozole model group was a successful PCOS model. Serum AMH and ovarian local expression of AMH and AMH-RII were both increased in letrozole model group. The elevated AMH had a positive correlation with T, growing follicle count and a negative correlation with body weight. CONCLUSIONS: The letrozole model group is a good animal model for the study of AMH in PCOS patients with obesity or insulin resistance. The increased serum AMH level in PCOS is the consequence of the androgen-induced excess of small antral follicles. These results lead to the hypothesis that reducing AMH may become a therapeutic target of PCOS, which is worth further research.


Asunto(s)
Hormona Antimülleriana/metabolismo , Inhibidores de la Aromatasa , Nitrilos , Ovario/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Triazoles , Animales , Hormona Antimülleriana/sangre , Modelos Animales de Enfermedad , Femenino , Letrozol , Folículo Ovárico/metabolismo , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/inducido químicamente , Ratas , Ratas Sprague-Dawley , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Testosterona/sangre
11.
Mol Hum Reprod ; 19(10): 676-86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23737337

RESUMEN

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.


Asunto(s)
Aborto Espontáneo/metabolismo , Quimiocinas CC/metabolismo , Decidua/citología , Receptores CCR10/metabolismo , Receptores CCR3/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo , Aborto Espontáneo/genética , Adulto , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células Cultivadas , Quimiocinas CC/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas In Vitro , Interleucina-17/farmacología , Interleucina-1beta/farmacología , Embarazo , Receptores CCR10/genética , Receptores CCR3/genética , Células del Estroma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Adulto Joven
12.
Am J Reprod Immunol ; 89(6): e13528, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35148017

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new type of coronavirus that has caused fatal infectious diseases and global spread. This novel coronavirus attacks target cells through the interaction of spike protein and angiotensin-converting enzyme II (ACE2), leading to different clinical symptoms. However, for a successful pregnancy, a well-established in-uterine environment includes a specific immune environment, and multi-interactions between specific cell types are prerequisites. The immune-related changes in patients infected with novel coronavirus could interfere with the immune microenvironment in the uterus, leading to fetal loss. We first reviewed the intrauterine environment in the normal development process and the possible pregnancy outcome in the infection state. Then, we summarized the immune response induced by SARS-CoV-2 in patients and analyzed the changes in ACE2 expression in the female reproductive system. Finally, the present observational evidence of infection in pregnant women was also reviewed.


Asunto(s)
COVID-19 , Humanos , Femenino , Embarazo , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2 , Peptidil-Dipeptidasa A/metabolismo , Resultado del Embarazo
13.
Elife ; 122023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36655976

RESUMEN

A defining feature of successful vaccination is the ability to induce long-lived antigen-specific memory cells. T follicular helper (Tfh) cells specialize in providing help to B cells in mounting protective humoral immunity in infection and after vaccination. Memory Tfh cells that retain the CXCR5 expression can confer protection through enhancing humoral response upon antigen re-exposure but how they are maintained is poorly understood. CXCR5+ memory Tfh cells in human blood are divided into Tfh1, Tfh2, and Tfh17 cells by the expression of chemokine receptors CXCR3 and CCR6 associated with Th1 and Th17, respectively. Here, we developed a new method to induce Tfh1, Tfh2, and Tfh17-like (iTfh1, iTfh2, and iTfh17) mouse cells in vitro. Although all three iTfh subsets efficiently support antibody responses in recipient mice with immediate immunization, iTfh17 cells are superior to iTfh1 and iTfh2 cells in supporting antibody response to a later immunization after extended resting in vivo to mimic memory maintenance. Notably, the counterpart human Tfh17 cells are selectively enriched in CCR7+ central memory Tfh cells with survival and proliferative advantages. Furthermore, the analysis of multiple human cohorts that received different vaccines for HBV, influenza virus, tetanus toxin or measles revealed that vaccine-specific Tfh17 cells outcompete Tfh1 or Tfh2 cells for the persistence in memory phase. Therefore, the complementary mouse and human results showing the advantage of Tfh17 cells in maintenance and memory function supports the notion that Tfh17-induced immunization might be preferable in vaccine development to confer long-term protection.


Asunto(s)
Memoria Inmunológica , Células T Auxiliares Foliculares , Humanos , Animales , Ratones , Células Th17/metabolismo , Linfocitos B , Linfocitos T Colaboradores-Inductores
14.
Clin Immunol ; 145(2): 161-73, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23069648

RESUMEN

The precise mechanism of characteristic Th2 predominance at maternal-fetal interface remains unresolved. In the present study, we investigated roles of the decidua-derived CCL2 in Th2 predominance at maternal-fetal interface. FCM shows that 55% CD56(+)CD16(-)CD3(-) decidual NK, 52% CD4(+) T cells and 75% CD14(+) monocytes express CCR2. Recombinant human CCL2 (rhCCL2) and the decidual stromal cells (DSCs)-derived supernatant can enhance proliferation and inhibit apoptosis of these decidual leukocytes (DLCs), and promote Th2 cytokines production, IL-4 and IL-10, with an increase in GATA-3 transcription. They also inhibit the secretion of Th1 cytokines, TNF-α and IFN-γ, with a decrease in T-bet transcription It is concluded that the secreted CCL2 by decidual stromal cells increases GATA-3 transcription and decreases T-bet transcription in the decidual leukocytes, which contributes to Th2 polarization at maternal-fetal interface. Furthermore, the Th2 cytokines, IL-4 and IL-10, rather than Th1 cytokines, was shown to increase CCL2 secretion of DSC.


Asunto(s)
Quimiocina CCL2/inmunología , Decidua/efectos de los fármacos , Células Asesinas Naturales/inmunología , Monocitos/inmunología , Embarazo/inmunología , Células del Estroma/metabolismo , Células Th2/inmunología , Adolescente , Adulto , Antígenos CD/genética , Antígenos CD/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Medios de Cultivo Condicionados , Decidua/citología , Decidua/inmunología , Femenino , Factor de Transcripción GATA3/agonistas , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Primer Trimestre del Embarazo , Receptores CCR2/genética , Receptores CCR2/inmunología , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Células del Estroma/citología , Células del Estroma/inmunología , Balance Th1 - Th2/efectos de los fármacos , Células Th2/citología , Células Th2/efectos de los fármacos , Transcripción Genética
15.
Blood ; 116(12): 2061-9, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20538796

RESUMEN

Thymic stromal lymphopoietins (TSLPs) play critical roles in dendritic cell-mediated immune responses. In this study, we found that human trophoblasts and decidual epithelial cells in maternal-fetal interface of early placentas express TSLP mRNA and protein, but only trophoblast cells secret soluble TSLP. Human decidual CD1c(+) DCs (dDCs) highly express the functional TSLP receptor complex TSLP receptor and interleukin-7 receptor-α. Recombinant human TSLP activates CD1C(+) decidual DCs and peripheral monocyte-derived DCs with increased costimulatory molecules, major histocompatibility complex class II, and OX-40L. Human TSLP or supernatants from human trophoblasts specifically stimulate dDCs to highly produce interleukin-10 and T(H)2-attracting chemokine CCL-17. The TSLP-activated dDCs prime decidual CD4(+) T cells for T(H)2 cell differentiation, involved in maternal-fetal immunotolerance. Interestingly, the protein expression of TSLP in normal pregnancy with significant T(H)2 bias is much higher than that of miscarriage showing T(H)1 bias at the maternal-fetal interface. Therefore, human trophoblasts may contribute to maternal-fetal tolerance by instructing dDCs to induce regulatory T(H)2 bias in human early pregnancy via TSLP.


Asunto(s)
Citocinas/inmunología , Células Dendríticas/inmunología , Células Th2/inmunología , Trofoblastos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Decidua/inmunología , Femenino , Feto/inmunología , Humanos , Tolerancia Inmunológica , Embarazo/inmunología , ARN Mensajero/análisis , Solubilidad , Trofoblastos/inmunología , Linfopoyetina del Estroma Tímico
16.
Endocrinology ; 163(11)2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36047434

RESUMEN

Endometrial decidualization refers to a series of morphological changes and functional remodeling of the uterine endometrium to accept the embryo under the effect of estrogen and progesterone secreted by ovaries after ovulation. During decidualization, endometrial stromal cells (ESCs) proliferate and differentiate into decidual stromal cells, undergoing cytoskeletal rearrangement-mediated morphological changes and expressing decidualization markers, such as insulin-like growth factor-binding protein-1 and prolactin. Ras homology (Rho) proteins, a family of small G proteins, are well known as regulators of cellular morphology and involved in multiple other cellular processes. In this study, we found ras homolog family member B (RHOB) was the most significantly upregulated gene in the Rho protein family after the in vitro decidualization of human primary ESCs. RhoB expression was induced mainly by 3',5'-cyclic adenosine 5'-monophosphate (cAMP) / protein kinase A (PKA) / cyclic adenosine monophosphate-response element binding protein signaling and partly by progesterone signaling. Knockdown of RhoB in ESCs greatly inhibited actin cytoskeletal rearrangement, cell morphological transformation, and upregulation of insulin-like growth factor-binding protein-1, suggesting an indispensable role of RhoB in decidualization. Mechanistically, the downstream target of RhoB was semaphorin3A (Sema3A), which mediated its signaling via interacting with the receptor, plexinA4. More importantly, decreased expression of RhoB, Sema3A, and plexinA4 were detected in deciduas from patients with unexplained spontaneous miscarriage. Collectively, our results indicate that RhoB/Sema3A/plexinA4 signaling plays a positive role in endometrial decidualization and relates to unexplained spontaneous miscarriage, which is worthy of further exploration so as to provide new insights into therapeutic strategies for pregnancy diseases associated with poor decidualization.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Receptores de Superficie Celular , Semaforina-3A , Células del Estroma , Proteína de Unión al GTP rhoB , Aborto Espontáneo/metabolismo , Actinas/metabolismo , Adenosina Monofosfato/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Decidua/metabolismo , Endometrio/metabolismo , Estrógenos/farmacología , Femenino , Humanos , Proteínas de Unión al GTP Monoméricas/metabolismo , Embarazo , Progesterona/metabolismo , Prolactina/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforina-3A/metabolismo , Células del Estroma/metabolismo , Proteína de Unión al GTP rhoB/metabolismo
17.
Asian J Androl ; 24(5): 441-444, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35532560

RESUMEN

During the coronavirus disease (COVID-19) epidemic, there have been concerns about the impact of vaccines on people's fertility, including the fertility of those who are currently preparing for pregnancy and those who might become pregnant in future. However, there is still a lack of research on the effect of the COVID-19 vaccine on male fertility, and it is not surprising that couples and donors have concerns regarding vaccination. In this study, a retrospective cohort study was conducted to examine semen quality before and after receipt of the inactivated COVID-19 vaccine. There were no statistically significant changes in semen parameters (volume, sperm concentration, progressive motility, and total progressive motile count) after two doses of vaccine (all P > 0.05). In summary, our study updates the most recent studies on the effects of the COVID-19 vaccine on male fertility, and the information from this study could be used to guide fertility recommendations for assisted reproductive technology (ART) patients and donors.


Asunto(s)
COVID-19 , Análisis de Semen , Vacunas contra la COVID-19 , Femenino , Humanos , Masculino , Embarazo , Estudios Retrospectivos , Semen , Recuento de Espermatozoides , Motilidad Espermática , Espermatozoides , Vacunación , Vacunas de Productos Inactivados
18.
Front Endocrinol (Lausanne) ; 13: 858176, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784569

RESUMEN

Background: Endometriosis (EMS), an endocrine-related inflammatory disease, is characterized by estrogen and progesterone imbalance in ectopic lesions. However, its pathogenic mechanism has not been fully elucidated. While SCM-198 is the synthetic form of leonurine and has multiple pharmacological activities such as antioxidation and anti-inflammation, it remains unknown whether it could inhibit the progress of EMS by regulating estrogen signaling and inflammation. Methods: The therapeutic effects of SCM-198 on EMS and its potential mechanism were analyzed by establishing EMS mouse models and performing an RNA sequencing (RNA-seq) assay. ELISA was performed to detect estrogen and tumor necrosis factor (TNF) -α concentrations in normal endometrial stromal cells (nESCs) and ectopic endometrial stromal cells (eESCs) with or without SCM-198 treatment. Western blotting, RNA silencing, and plasmid overexpression were used to analyze the relationship between inflammation, endocrine factors, and autophagy and the regulatory activity of SCM-198 on the inflammation-endocrine-autophagy axis. Results: Increased estrogen-estrogen receptor (ER) α signaling and decreased progesterone receptor isoform B (PRB) expression synergistically led to a hypo-autophagy state in eESCs, which further inhibited the apoptosis of eESCs. The high expression of TNF-α in eESCs enhanced the antiapoptotic effect mediated by low autophagy through the activation of the aromatase-estrogen-ERα signaling pathway. SCM-198 inhibited the growth of ectopic lesions in EMS mice and promoted the apoptosis of eESCs both in vivo and in vitro. The apoptotic effect of SCM-198 on eESCs was attained by upregulating the autophagy level via the inhibition of the TNF-α-activated aromatase-estrogen-ERα signal and the increase in PRB expression. Conclusion: Inflammation facilitated the progress of EMS by disrupting the estrogen regulatory axis. SCM-198 inhibited EMS progression by regulating the inflammation-endocrine-autophagy axis.


Asunto(s)
Endometriosis , Animales , Aromatasa/genética , Aromatasa/metabolismo , Autofagia , Endometriosis/metabolismo , Endometriosis/prevención & control , Endometrio/patología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Ácido Gálico/análogos & derivados , Humanos , Ratones , Receptores de Progesterona/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Factor de Necrosis Tumoral alfa/metabolismo
19.
Int J Biol Sci ; 18(5): 1961-1973, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35342349

RESUMEN

Background: Endometriosis (EMS), a typical endocrine immune disorder, associates with dramatically increased estrogen production and disorganized immune response in ectopic focus. Peritoneal regulatory T cells (Tregs) expansion in women with EMS and their pathogenic role attributable to endometriotic immunotolerance has been reported. Whether local high estrogen promotes EMS by discipling Tregs needs to be further explored. Up to date, there is no effective medicine for the treatment of EMS. SCM-198 is a synthetic leonurine with multiple physiological activities. Whether SCM-198 could regulate Tregs via estrogen and facilitate the radical cure of EMS has not yet been reported. Methods: Proportion of Tregs in peritoneal fluid of patients with EMS was firstly analyzed via flow cytometry. Peritoneal estrogen concentration and the mRNA levels of estrogen receptor α (ERα) and estrogen receptor ß (ERß) of Tregs were detected by ELISA and RT-PCR, respectively. Grouped in vitro induction assays were performed to explore the effects of SCM-198 and estrogen signaling on Tregs. Cell invasion and viability assays were utilized to detect the crosstalk between Tregs and ectopic endometrial stromal cells (eESCs), with or without SCM-198 treatment. Furthermore, EMS mice models were established to verify the therapeutic effects of SCM-198. Results: Increased Tregs were found in peritoneal fluid of EMS patients, accompanied with estrogen-ERα overactivation. Estrogen-ERα triggered the expansion of Tregs and their cytokine production (IL-10 and TGF-ß1), which could be reversed by SCM-198 treatment. Moreover, SCM-198 abated the invasion and viability of eESCs enhanced by Tregs. In vivo experiments confirmed that SCM-198 obviously retarded the growth of ectopic lesions and downregulated the functions of Tregs via estrogen-ERα inactivation. Conclusions: These data suggest that SCM-198 attenuates Tregs expansion via the inhibition of estrogen-ERα signaling in EMS and offer a promising therapy for such a refractory disease.


Asunto(s)
Endometriosis , Receptor alfa de Estrógeno , Animales , Endometriosis/tratamiento farmacológico , Endometriosis/genética , Receptor alfa de Estrógeno/genética , Estrógenos , Femenino , Ácido Gálico/análogos & derivados , Humanos , Ratones , Linfocitos T Reguladores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA