Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cancer Res Clin Oncol ; 147(1): 183-194, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32995956

RESUMO

PURPOSE: BRAFV600E, a major driver of thyroid cancer, evaluated in the context of thyroid hormones and human relaxin. METHODS: Immunohistochemical expressions of BRAFV600E, TSH, TSH receptor (TSHR), T4, T3 receptor (T3R), RLNH2, and its receptor, RXFP1, were evaluated in thyroid tumors from a retrospective U.S. population of 481 cancer cases diagnosed in 1983-2004. RESULTS: BRAFV600E was expressed in 52% of all thyroid tumors; expression of other markers ranged from 25% for T4 to 98% for RLNH2. Tumors predominantly exhibited hypothyroid-like conditions characterized by elevated TSH and TSHR and reduced T4. BRAFV600E prevalence was significantly higher in tumors expressing TSH, TSHR, T3R, and RXFP1 and lower in tumors expressing T4. The proportion of BRAFV600E mutation in classic papillary tumors significantly increased from 56 to 72% over the 21-year period of diagnoses, while expression of RXFP1, TSH, TSHR, and T3R decreased in non-tumor. Racial/ethnic differences were observed in thyroid hormone marker expression. Non-tumor expression of TSH, TSHR, and T3R were each associated with shorter overall survival, but did not remain significant after adjustment for demographic and clinical factors. CONCLUSIONS: Our study provides the first evidence of the potential interaction of BRAFV600E mutation, relaxin, and thyroid hormones in thyroid carcinogenesis. Moreover, our results suggest that hypothyroidism, influenced by RLNH2 activity, may underlie the development of the majority of thyroid cancers and mediate the role of BRAFV600E in thyroid carcinogenesis. BRAFV600E mutation is increasing in papillary thyroid cancers and may be contributing to the rising incidence of this malignancy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Papilar/patologia , Hipotireoidismo/fisiopatologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Relaxina/metabolismo , Neoplasias da Glândula Tireoide/patologia , Idoso , Biomarcadores Tumorais/genética , Carcinoma Papilar/genética , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Relaxina/genética , Estudos Retrospectivos , Taxa de Sobrevida , Neoplasias da Glândula Tireoide/genética
2.
Reproduction ; 154(1): 67-77, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28468839

RESUMO

Relaxin, a systemic and placental hormone, has potential roles in fetoplacental growth. Human placenta expresses two RLN genes, RLNH1 and RLNH2 Maternal obesity is common and is associated with abnormal fetal growth. Our aims were to relate systemic and cord blood RLNH2, placental RLNs and their receptor (RXFP1) with fetoplacental growth in context of maternal body mass index, and associations with insulin-like growth factor 2 (IGF2) and vascular endothelial growth factor A (VEGFA) in the same placentas. Systemic, cord blood and placental samples were collected prior to term labor, divided by prepregnancy body mass index: underweight/normal (N = 25) and overweight/obese (N = 44). Blood RLNH2 was measured by ELISA; placental RLNH2, RLNH1, RXFP1, IGF2 and VEGFA were measured by quantitative immunohistochemistry and mRNAs were measured by quantitative reverse transcription PCR. Birthweight increased with systemic RLNH2 only in underweight/normal women (P = 0.036). Syncytiotrophoblast RLNH2 was increased in overweight/obese patients (P = 0.017) and was associated with placental weight in all subjects (P = 0.038). RLNH1 had no associations with birthweight or placental weight, but was associated with increased trophoblast and endothelial IGF2 and VEGFA, due to female fetal sex. Thus, while systemic RLNH2 may be involved in birthweight regulation in underweight/normal women, placental RLNH2 in all subjects may be involved in placental weight. A strong association of trophoblast IGF2 with birthweight and placental weight in overweight/obese women suggests its importance. However, an association of only RLNH1 with placental IGF2 and VEGFA was dependent upon female fetal sex. These results suggest that both systemic and placental RLNs may be associated with fetoplacental growth.


Assuntos
Desenvolvimento Fetal/fisiologia , Insulina/fisiologia , Placenta/fisiologia , Proteínas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/fisiologia , Peso ao Nascer , Índice de Massa Corporal , Feminino , Sangue Fetal/química , Feto , Expressão Gênica , Humanos , Imuno-Histoquímica , Insulina/análise , Insulina/sangue , Fator de Crescimento Insulin-Like II/análise , Obesidade/complicações , Obesidade/fisiopatologia , Tamanho do Órgão , Placenta/química , Placenta/patologia , Gravidez , Complicações na Gravidez/fisiopatologia , Proteínas/análise , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/sangue , Receptores de Peptídeos/análise , Receptores de Peptídeos/sangue , Fatores Sexuais , Fator A de Crescimento do Endotélio Vascular/análise
3.
Placenta ; 41: 53-61, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27208408

RESUMO

INTRODUCTION: α-klotho is an anti-aging protein, potentially important in preeclampsia (PE). Produced by kidney, brain and placenta, and by mRNA splicing is both a full-length membrane-bound and a truncated soluble protein in the circulation. The membrane-bound protein is an obligate co-receptor for fibroblast growth factor 23 (FGF23) and its action on receptor (FGFR), but ADAM proteinases also cause its shedding. The aims of this study were to investigate levels of maternal plasma, placental, and fetal membrane α-Klotho and their association with placental accelerated villous maturation (AVM) in PE. In addition, placental and membrane levels of ADAM17 and FGFR were measured in the same patients. METHODS: Maternal blood, placenta and fetal membranes from 61 women (31 with PE and 30 controls) between 32 and 40 weeks gestation were collected. Plasma α-klotho was measured by ELISA, and quantitative immunohistochemistry used for α-klotho, ADAM17 and FGFR1 in tissues. Placental AVM was histologically assessed. RESULTS: Maternal plasma levels of α-Klotho were higher in PE compared to controls (p = 0.01) and patients with the highest levels had significantly less AVM (p = 0.03). α-Klotho, ADAM17, and FGFR were all present in syncytiotrophoblast and cytotrophoblast of membranes. Between 32 and 40 weeks gestation, all placental levels decreased in controls respectively (p = 0.04, p = 0.004, p = 0.05), but not in PE. Fetal membrane levels were unchanged. DISCUSSION: Maternal plasma α-Klotho was increased in PE and its levels associated with reduced placental AVM. Changes in placental α-Klotho, ADAM17, and FGFR suggest their involvement in the pathophysiology of PE.


Assuntos
Idade Gestacional , Glucuronidase/análise , Placenta/química , Pré-Eclâmpsia/fisiopatologia , Proteína ADAM17/análise , Adulto , Membranas Extraembrionárias/química , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/sangue , Humanos , Proteínas Klotho , Gravidez , Terceiro Trimestre da Gravidez , Receptores de Fatores de Crescimento de Fibroblastos/análise , Trofoblastos/química
4.
Reprod Sci ; 22(8): 1028-36, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25670718

RESUMO

Visfatin is both a systemic adipocytokine and the cytosolic enzyme, nicotinamide phosphoribosyl transferase (Nampt). This is a longevity protein, which extends the lifespan of human cells by activating sirtuin 1 (SIRT1). In this study, we sought a role for these proteins in obese pregnant women, who experience more postterm deliveries. Thus, 78 women (26 lean, 24 overweight, and 28 obese) were recruited and maternal blood and placental tissue collected prior to term labor. Plasma levels were measured by enzyme-linked immunosorbent assay and quantitative immunohistochemistry used for placenta. We confirmed maternal plasma interleukin 6 increased according to prepregnancy body mass index (BMI; P < .0001) and showed a linear relationship between BMI and syncytiotrophoblast visfatin/Nampt (P = .021) but not with its levels in maternal plasma. Both systemic and placental visfatin/Nampt were significantly associated with placental SIRT1 levels (P = .028 and .017). Thus, higher visfatin/Nampt may prevent a labor-associated decrease in SIRT1 leading to postterm delivery in obesity.


Assuntos
Citocinas/sangue , Criança Pós-Termo , Nicotinamida Fosforribosiltransferase/sangue , Obesidade/complicações , Placenta/química , Complicações na Gravidez/etiologia , Sirtuína 1/sangue , Adulto , Biomarcadores/sangue , Índice de Massa Corporal , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Feminino , Idade Gestacional , Humanos , Imuno-Histoquímica , Interleucina-6/sangue , Modelos Lineares , Obesidade/sangue , Obesidade/diagnóstico , Gravidez , Complicações na Gravidez/sangue , Fatores de Risco
5.
Reprod Sci ; 22(2): 189-97, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24899470

RESUMO

This study aimed to gain new insights into both systemic and placental leptin and its receptors, with reference to the maternal prepregnancy body mass index (BMI). Thus, 84 women (29 lean, 24 overweight, and 31 obese) were recruited and maternal, cord blood, and placental tissues collected prior to term labor. Plasma levels were measured by enzyme-linked immunosorbent assay and for placenta, immunohistochemistry and messenger RNAs (mRNAs) were quantitated. We confirmed that maternal leptin increased linearly as the soluble receptor decreased with BMI (P = .001). Fetal leptin increased with maternal BMI (P = .02) and birth weight (P = .006) and was higher in female infants (P < .001). Placental mRNA levels of leptin and its receptors showed no change in BMI. However, we show a significant (P = .043) linear increase in leptin in the placental vascular endothelial cells with maternal obesity, while leptin in syncytiotrophoblast showed no statistical change. Leptin receptors localized to syncytiotrophoblast and intravillous macrophages and were unchanged with BMI.


Assuntos
Leptina/sangue , Obesidade/sangue , Placenta/química , Receptores para Leptina/análise , Adulto , Biomarcadores/sangue , Peso ao Nascer , Índice de Massa Corporal , Células Endoteliais/química , Ensaio de Imunoadsorção Enzimática , Feminino , Sangue Fetal/química , Humanos , Imuno-Histoquímica , Recém-Nascido , Leptina/genética , Macrófagos/química , Masculino , Obesidade/diagnóstico , Obesidade/genética , Gravidez , RNA Mensageiro/análise , Reação em Cadeia da Polimerase em Tempo Real , Receptores para Leptina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Trofoblastos/química
6.
Ann N Y Acad Sci ; 1160: 60-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19416160

RESUMO

The LGR7/RXFP1 and LGR8/RXFP2 receptors are unique receptors among the G-protein-coupled receptors (GPCRs) in having a low-density lipoprotein class A (LDL-A) module. Their complex gene organization, among the intron-richest of the GPCRs, suggests that alternative splicing is a common occurrence. We have therefore investigated the role of the LDL-A module and shown the identity, expression, and functions of three LGR7 splice variants in the human decidua. Point mutations of conserved residues or complete deletion of the LDL-A module resulted in loss of the cAMP response to relaxin. Its glycosylation also impacted LGR7 cell surface delivery and therefore receptor activation. The wild-type (WT) LGR7 was expressed as both precursor and mature forms, but deletion of the LDL-A module resulted in expression of only the mature form. Three new alternatively spliced variants of LGR7 were identified, all containing a truncated extracellular region. Their functional characterization showed them exerting dominant negative effects on the WT LGR7 by preventing its homodimerization, maturation, and subsequent trafficking to the cell surface, resulting in loss of function. In summary, different mechanisms have been identified for controlling the cell surface expression and function of the LGR7 protein which are likely to be significant for the role of relaxin in human parturition.


Assuntos
Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/fisiologia , Linhagem Celular , AMP Cíclico/metabolismo , Humanos , Mutação Puntual , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Relaxina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Relação Estrutura-Atividade
8.
Endocrinology ; 148(3): 1181-94, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17158203

RESUMO

The relaxin receptor (LGR7, relaxin family peptide receptor 1) is a member of the leucine-rich repeat containing G protein-coupled receptors subgroup C. This and the LGR8 (relaxin family peptide receptor 2) receptor are unique in having a low-density lipoprotein class A (LDL-A) module at their N termini. This study was designed to show the role of the LDL-A in LGR7 expression and function. Point mutants for the conserved cysteines (Cys(47) and Cys(53)) and for calcium binding asparagine (Asp(58)), a mutant with deleted LDL-A domain and chimeric LGR7 receptor with LGR8 LDL-A all showed no cAMP response to human relaxins H1 or H2. We have shown that their cell surface delivery was uncompromised. The mutation of the putative N-linked glycosylation site (Asn(36)) decreased cAMP production and reduced cell surface expression to 37% of the wild-type LGR7. All point mutant, chimeric, and wild-type receptor proteins were expressed as the two forms. The immature or precursor form of the receptor was 80 kDa, whereas the mature receptor, delivered to the cell surface was 95 kDa. The glycosylation mutant was also expressed as two forms with appropriately smaller molecular masses. Deletion of the LDL-A module resulted in expression of the mature receptor only. These data suggest that the LDL-A module of LGR7 influences receptor maturation, cell surface expression, and relaxin-activated signal transduction.


Assuntos
Membrana Celular/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Sequência de Aminoácidos , Células Cultivadas , Deleção de Genes , Glicosídeo Hidrolases/farmacologia , Humanos , Proteínas Relacionadas a Receptor de LDL/química , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteínas Relacionadas a Receptor de LDL/fisiologia , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/fisiologia , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico , Receptores de Peptídeos , Homologia de Sequência de Aminoácidos , Transdução de Sinais
9.
Arthritis Rheum ; 54(7): 2084-95, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16802343

RESUMO

OBJECTIVE: To determine whether interleukin-6 (IL-6) trans-signaling directs the expression of pre-B cell colony-enhancing factor (PBEF) in vitro and in vivo. METHODS: Complementary DNA from rheumatoid arthritis (RA) synovial fibroblasts treated with IL-6 and soluble IL-6 receptor (sIL-6R) was used to probe a cytokine microarray. PBEF regulation by the IL-6-related cytokines, IL-6, sIL-6R, oncostatin M (OSM), IL-11, and leukemia inhibitory factor (LIF) was determined by reverse transcription-polymerase chain reaction analysis. IL-6-mediated STAT-3 regulation of PBEF was determined using a cell-permeable STAT-3 inhibitor peptide. Antigen-induced arthritis (AIA) was induced in wild-type (IL-6(+/+)) and IL-6-deficient (IL-6(-/-)) mice. PBEF and STAT were detected by immunohistochemistry, immunoblotting, and electrophoretic mobility shift assay. Synovial levels of PBEF were quantified by enzyme immunoassay. RESULTS: IL-6 trans-signaling regulated PBEF in a STAT-3-dependent manner. In addition, PBEF was regulated by the IL-6-related cytokine OSM, but not IL-11 or LIF. Flow cytometric analysis of the IL-6-related cognate receptors suggested that OSM regulates PBEF via its OSM receptor beta and not its LIF receptor. The involvement of PBEF in arthritis progression was confirmed in vivo, where induction of AIA resulted in a 4-fold increase in the synovial expression of PBEF. In contrast, little or no change was observed in IL-6(-/-) mice, in which the inflammatory infiltrate was markedly reduced and synovial STAT-1/3 activity was also impaired. Analysis of human RA synovial tissue confirmed that PBEF immunolocalized in apical synovial membrane cells, endothelial cells, adipocytes, and lymphoid aggregates. Synovial fluid levels of PBEF were significantly higher in RA patients than in osteoarthritis patients. CONCLUSION: Experiments presented herein demonstrate that PBEF is regulated via IL-6 trans-signaling and the IL-6-related cytokine OSM. PBEF is also actively expressed during arthritis. Although these data confirm an involvement of PBEF in disease progression, the consequence of its action remains to be determined.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/fisiopatologia , Citocinas/metabolismo , Interleucina-6/fisiologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Citocinas/genética , Citocinas/farmacologia , Citocinas/fisiologia , Progressão da Doença , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-11/farmacologia , Interleucina-6/genética , Interleucina-6/farmacologia , Fator Inibidor de Leucemia , Camundongos , Camundongos Knockout , Nicotinamida Fosforribosiltransferase , Análise de Sequência com Séries de Oligonucleotídeos , Oncostatina M , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/fisiologia , Fator de Transcrição STAT3/genética , Transdução de Sinais/genética , Líquido Sinovial/efeitos dos fármacos , Líquido Sinovial/metabolismo , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
10.
Am J Obstet Gynecol ; 193(1): 273-82, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16021090

RESUMO

OBJECTIVE: The purpose of this study was to determine whether pre-B-cell colony-enhancing factor is a secreted cytokine in the human amnion and to study its chemotaxic and antiapoptotic properties. STUDY DESIGN: Pre-B-cell colony-enhancing factor secretion was studied from amniotic epithelial-like WISH cells and primary amniotic epithelial cells that were seeded on squares of immobilon-P membrane and stimulated with lipopolysaccharide or tumor necrosis factor-alpha, respectively. The pre-B-cell colony-enhancing factor protein was detected both intracellularly and after secretion, as bound to the membrane, by immunostaining and densitometry. Medium and cell lysates that were obtained from WISH cells that were treated with lipopolysaccharide alone or together with a pre-B-cell colony-enhancing factor antisense oligonucleotide to block pre-B-cell colony-enhancing factor translation were also analyzed for secreted pre-B-cell colony-enhancing factor by Western blotting and densitometry. A chemotaxic effect of pre-B-cell colony-enhancing factor on human neutrophils was compared with the chemoattractants interleukin-8 and N-Formyl-Met-Leu-Phe methyl ester in a rapid fluorescence-based neutrophil migration assay. Apoptosis was induced in primary amniotic epithelial cells and fibroblasts by actinomycin D (1 microg/mL); the antiapoptotic effects of pre-B-cell colony-enhancing factor on early apoptosis were measured by the annexin V assay, and the late effects were determined by measurement of nuclear matrix protein in the media. RESULTS: Treatment of amnion cells that adhered to immobilon-P membrane to induce the secretion of pre-B-cell colony-enhancing factor showed significantly (P<.05) more pre-B-cell colony-enhancing factor protein surrounding the cells compared with the controls. Although the addition of lipopolysaccharide to cultured WISH cells caused the secretion of pre-B-cell colony-enhancing factor into the medium, co-treatment with an antisense oligonucleotide to pre-B-cell colony-enhancing factor obliterated it. Analysis of the cell lysates showed no significant change, which suggests that most of the pre-B-cell colony-enhancing factor protein had been secreted. No significant chemotaxic effects of pre-B-cell colony-enhancing factor were observed; however, pre-B-cell colony-enhancing factor treatment (100 ng/mL), together with actinomycin D, cancelled the early induction of apoptosis, although there was a dose-dependent and significant late antiapoptotic effect on primary amnion epithelial cells (P<.001) and fibroblasts (P<.01). CONCLUSION: Pre-B-cell colony-enhancing factor is a secreted protein from amniotic epithelial cells. Although it had no chemotaxic effects, it was antiapoptotic for both amniotic epithelial cells and fibroblasts and may protect these cells against apoptosis that is induced by chronic distension, labor, or infection.


Assuntos
Âmnio/metabolismo , Citocinas/metabolismo , Âmnio/citologia , Âmnio/fisiologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular , Quimiotaxia de Leucócito/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/genética , Citocinas/farmacologia , Células Epiteliais/fisiologia , Epitélio/metabolismo , Feminino , Fibroblastos/fisiologia , Humanos , Neutrófilos/fisiologia , Nicotinamida Fosforribosiltransferase , Oligorribonucleotídeos Antissenso/farmacologia , Gravidez , Proteínas Recombinantes/farmacologia
11.
Mol Cell Endocrinol ; 219(1-2): 115-25, 2004 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-15149733

RESUMO

Relaxin, a peptide hormone important to the outcome of human pregnancy is expressed in a tissue specific manner as two genes known as relaxins H1 and H2, in addition to a third human relaxin H3, expressed primarily in the brain. The H1 and H2 genes are highly homologous, differentially expressed in reproductive tissues and appear to activate the same receptor, but their regulation is poorly understood. Based upon the known physiology of these hormones and the response elements in their 5'- and 3'-flanking regions, the possibility that progesterone and/or the glucocorticoids might influence their differential expression was therefore investigated. The changes in the mRNA levels of the relaxin genes in response to either medroxyprogesterone acetate (MPA) or dexamethasone (Dex) were analyzed by RT-PCR using a choriocarcinoma cell line (JAR) as a model system, because the expression of these genes in any primary human cell type is too low for such a study. The addition of 0.5 microM MPA to JAR cells, significantly upregulated the mRNA of only the relaxin H2, while the addition of 0.5 microM Dex significantly upregulated the mRNAs for both the relaxins, after 6h of treatment. Promoter assays indicated an early activation of transcription (1 h), which by 6 h had decreased. Progesterone and/or glucocorticoids could exert their effects via the GRE motif found on the 5'-flanking region of the relaxin genes. The H1-GRE differs from the H2-GRE by a single nucleotide, which may affect H1-GRE binding to the progesterone receptor (PR) but not the glucocorticoid receptor (GR). The antiprogestin RU486 inhibited the binding of the GR to both H1-GRE and H2-GRE, while it enhanced the binding of the PR to these GREs. As determined by gel shift assays, this GRE motif could bind to both the PR and GR and was therefore considered to be functional. Thus, both progesterone and glucocorticoids are capable of differentially regulating the expression of the two human relaxin genes in a model system.


Assuntos
Glucocorticoides/farmacologia , Progesterona/farmacologia , Relaxina/genética , Motivos de Aminoácidos , Dexametasona/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica , Humanos , Acetato de Medroxiprogesterona/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica , RNA Mensageiro/análise
12.
Am J Obstet Gynecol ; 189(4): 1187-95, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14586377

RESUMO

OBJECTIVE: Our purpose was to show the effects of pre-B-cell colony-enhancing factor on the genes that are expressed by the human fetal membranes. STUDY DESIGN: Explants of fetal membranes (amnion, chorion, and decidua) from three term patients were treated with 100 ng/mL recombinant human pre-B-cell colony-enhancing factor for 4 hours. RNAs were hybridized to gene chips that contained >18,000 known genes. One experiment was done in triplicate to assess replication. Data were analyzed to quantitate the signal intensities of each complementary DNA on the array. Confirmation of the results was carried out on tissues from nine other patients by the measurement of the proteins or quantitative real-time reverse transcriptase-polymerase chain reaction. RESULTS: Replication gave <92.6% identical results, which showed high method reproducibility. Pre-B-cell colony-enhancing factor treatment caused a significant increase in 103 genes and decrease in 139 genes. Only 8 genes were up-regulated consistently and significantly in all three patients (three key inflammatory cytokines [tumor necrosis factor-alpha, interleukin-6, and interleukin-1beta], four important chemokines [macrophage inflammatory protein-1alpha, macrophage inflammatory protein-1beta, macrophage inflammatory protein-3alpha, and growth-related oncogene-gamma], and prostaglandin-endoperoxide synthase 2). These data were confirmed by the measurement in the media with the use of specific enzyme-linked immunosorbent assays for tumor necrosis factor-alpha, interleukin-6, and interleukin-1beta, macrophage inflammatory protein-1alpha, macrophage inflammatory protein-1beta, and macrophage inflammatory protein-3alpha and by quantitative real-time reverse transcriptase-polymerase chain reaction for growth-related oncogene-gamma and prostaglandin-endoperoxide synthase 2. CONCLUSION: Pre-B-cell colony-enhancing factor appears to be at the proximal end of the pathway to labor initiation and may link sterile distention-induced labor with that of infection-induced labor.


Assuntos
Citocinas/fisiologia , Membranas Extraembrionárias/fisiologia , Análise Serial de Proteínas , Âmnio/efeitos dos fármacos , Quimiocinas/análise , Córion/efeitos dos fármacos , Citocinas/análise , Citocinas/genética , Citocinas/farmacologia , Decídua/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Membranas Extraembrionárias/efeitos dos fármacos , Feminino , Humanos , Técnicas In Vitro , Nicotinamida Fosforribosiltransferase , Reação em Cadeia da Polimerase , Gravidez , Proteínas Recombinantes/farmacologia
13.
Am J Obstet Gynecol ; 188(1): 234-41, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12548223

RESUMO

OBJECTIVE: The study was conducted to determine whether relaxin has a proliferative effect on amniotic epithelial cells and to show that this effect is caused by its stimulation of the insulin-like growth factor-II (IGF-II) gene. STUDY DESIGN: Immunolocalization and Northern analysis were used to confirm the expression of IGF-II by the fetal cells in the membranes. Human amniotic epithelial (WISH) cells were treated with doses of IGF-II or human relaxin and their proliferative effects measured. The mechanism of the effect of relaxin on cellular proliferation was studied with the use of an IGF-II-blocking antibody and Northern analysis for IGF-II gene expression after treatment with relaxin. An in vivo correlate was sought by quantitation of relaxin gene expression in 10 fetal membranes from women with normally grown and large for gestational age infants. RESULTS: The amniotic epithelial and cytotrophoblast cells of the fetal membranes expressed IGF-II, as did the amniotic epithelial-like (WISH) cell line. Treatment of WISH cells with IGF-II or relaxin caused a significant (P <.03) and dose-related increase in WISH cell proliferation over 5 days. The concurrent treatment with a blocking antibody to IGF-II significantly decreased the proliferative response to IGF-II (P <.002) and relaxin (P <.002). Treatment with relaxin caused a significant increase (P <.003) in the transcription of IGF-II in 24 hours. In fetal membranes, the levels of relaxin gene expression correlated with fetal membrane surface area (r = 0.76) and was significantly greater (P <.008) in the membranes from macrosomic infants (4020-4729 g) compared with those normally grown (2855-3830 g). CONCLUSION: IGF-II and relaxin both caused the proliferation of WISH cells. Concurrent treatment with an IGF-II-blocking antibody abrogated the proliferative effects of both hormones. Relaxin increased the transcription of IGF-II, and its expression levels in the fetal membranes correlated with the membrane surface area as well as neonatal birth weight. These data suggest that relaxin is a growth factor for the fetal membranes.


Assuntos
Âmnio/citologia , Divisão Celular/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/fisiologia , Relaxina/farmacologia , Âmnio/química , Anticorpos/farmacologia , Northern Blotting , Células Cultivadas , Córion/química , Decídua/química , Células Epiteliais/química , Células Epiteliais/citologia , Feminino , Macrossomia Fetal/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Imunoensaio , Fator de Crescimento Insulin-Like II/análise , Fator de Crescimento Insulin-Like II/genética , Placenta/química , Gravidez , RNA Mensageiro/análise , Relaxina/genética
14.
Am J Obstet Gynecol ; 187(4): 1051-8, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12389004

RESUMO

OBJECTIVE: Our purpose was to determine whether pre-B-cell colony-enhancing factor (PBEF) is expressed in the human fetal membranes during normal gestation and parturition in the absence of infection and to show its effects on the expression of interleukin (IL)-6 and IL-8. STUDY DESIGN: PBEF was immunolocalized in the fetal membranes from early pregnancy, at preterm, and at term. Its expression was quantitated by Northern analysis in separated uninfected amnion, chorion, decidua, and placenta of patients at term before labor and in full-thickness membranes before and after spontaneous labor at preterm and at term. Amnion-like epithelial (WISH) cells and fetal membrane explants were treated with recombinant PBEF (rhPBEF), and the expression of IL-6 and IL-8 was quantitated. RESULTS: PBEF was immunolocalized throughout gestation in the amniotic epithelium and mesenchymal cells as well as the chorionic cytotrophoblast and parietal decidua. Northern analysis showed significantly more (P <.01) PBEF expressed in the amnion than in either chorion or placenta. Its expression increased after labor at both preterm and term and correlated with that of IL-8 (r = 0.87). rhPBEF treatment of WISH cells significantly increased IL-6 (P <.05) and IL-8 (P <.01) gene expression after 4 hours and of IL-8 protein after 24 hours (P <.01); similar 4-hour treatment of fetal membrane explants significantly increased IL-6 (P <.01) and IL-8 (P <.05) gene expression. CONCLUSION: PBEF is a novel cytokine constitutively expressed by the fetal membranes during pregnancy. It increased the expression of IL-6 and IL-8 and may be important in both normal spontaneous labor and infection-induced preterm labor.


Assuntos
Âmnio/metabolismo , Córion/metabolismo , Citocinas/metabolismo , Âmnio/efeitos dos fármacos , Northern Blotting , Linhagem Celular , Córion/efeitos dos fármacos , Citocinas/farmacologia , Decídua/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Técnicas Imunológicas , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Trabalho de Parto/metabolismo , Nicotinamida Fosforribosiltransferase , Trabalho de Parto Prematuro/metabolismo , Placenta/metabolismo , Gravidez , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA