Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Lupus ; 22(6): 583-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23535532

ABSTRACT

OBJECTIVE: Systemic lupus erythematosus (SLE) is an autoimmune disease that affects women nine times more often than men. The present study investigates estradiol-dependent control of the calcium-buffering protein, calreticulin, to gain further insight into the molecular basis of abnormal T cell signaling in SLE T cells. METHODS: T cells were purified from blood samples obtained from healthy females and SLE patients. Calreticulin expression was quantified by real-time polymerase chain amplification. Calreticulin and estrogen receptor-α were co-precipitated and analyzed by Western blotting to determine if the proteins associate in T cells. RESULTS: Calreticulin expression increased (p = 0.034) in activated control T cells, while estradiol decreased (p = 0.044) calreticulin in resting T cells. Calreticulin expression decreased in activated SLE T cell samples and increased in approximately 50% of resting T cell samples. Plasma estradiol was similar (p > 0.05) among SLE patients and control volunteers. Estrogen receptor-α and calreticulin co-precipitated from nuclear and cytoplasmic T cell compartments. CONCLUSIONS: The results indicate that estradiol tightly regulates calreticulin expression in normal human T cells, and the dynamics are different between activated and resting T cells. The absence of this tight regulation in SLE T cells could contribute to abnormal T cell function.


Subject(s)
Calreticulin/metabolism , Estradiol/metabolism , Lupus Erythematosus, Systemic/physiopathology , T-Lymphocytes/pathology , Adult , Blotting, Western , Case-Control Studies , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Humans , Lupus Erythematosus, Systemic/epidemiology , Middle Aged , Real-Time Polymerase Chain Reaction , Sex Factors , Signal Transduction , T-Lymphocytes/metabolism
2.
Lupus ; 17(6): 548-54, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18539708

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease that occurs primarily in women of reproductive age. The disease is characterized by exaggerated T-cell activity and abnormal T-cell signalling. The mitogen-activated protein kinase (MAPK) pathway is involved in the maintenance of T-cell tolerance that fails in patients with SLE. Oestrogen is a female sex hormone that binds to nuclear receptors and alters the rate of gene transcription. Oestrogen can also act through the plasma membrane and rapidly stimulate second messengers including calcium flux and kinase activation. In this study, we investigated whether oestrogen influences the activation of MAPK signalling through the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in activated SLE T cells. SLE and control T cells were cultured in serum-free medium without and with oestradiol (10(-7) M) for 18 h. The T cells were activated with phorbol 12 myristate 13-acetate and ionomycin for various time points (0-60 min), and the amount of phosphorylated ERK1/2 was measured by immunoblotting. There were no differences in ERK1/2 phosphorylation between SLE and control T cells at 5 and 15 min after the activation stimulus. However, comparison between the amount of phosphorylated ERK1/2 in SLE T cells from the same patients cultured without and with oestradiol showed a significant oestrogen-dependent suppression (P=0.48) of ERK1/2 in patients with inactive/mild systemic lupus erythematosus disease activity index (SLEDAI) (0-2) compared with patients with moderate (4-6) or active (8-12) SLEDAI scores. These results suggest that the suppression of MAPK through ERK1/2 phosphorylation is sensitive to oestradiol in patients with inactive or mild disease, but the sensitivity is not maintained when disease activity increases. Furthermore, studies are now necessary to understand the mechanisms by which oestrogen influences MAPK activation in SLE T cells.


Subject(s)
Estrogens/metabolism , Lupus Erythematosus, Systemic/metabolism , MAP Kinase Signaling System , Adult , Cells, Cultured , Enzyme Activation , Female , Humans , Lupus Erythematosus, Systemic/physiopathology , Lymphocyte Activation , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology
3.
Lupus ; 15(12): 852-7, 2006.
Article in English | MEDLINE | ID: mdl-17211990

ABSTRACT

Previous studies in our laboratory showed a dose-dependent and hormone-specific increase in CD154 expression in T cells from females with systemic lupus erythematosus (SLE). This present study investigates if the estrogen-dependent increase in CD154 expression is due to stabilization of the messenger RNA. T cells from female SLE patients and controls were cultured for 18 h in serum-free medium without and with estradiol 17-beta (10(-7) M). T cells were either unstimulated (resting) or were activated by further culture on anti-CD3 coated plates. Actinomycin D (25 microg/mL) was added to parallel cultures to inhibit new messenger RNA synthesis. CD154 messenger RNA stability was assessed by reverse-transcription polymerase chain amplification. Resting SLE (n = 10, P = 0.88) and normal (n = 7, P = 0.65) T cells showed no significant differences in message stability in response to estradiol. CD154 messenger RNA was also not significantly stabilized in activated SLE (n = 10, P = 0.15) or activated normal (n = 6, P = 0.077) T cells in response to estradiol. These findings indicate that the estrogen-dependent increase in CD154 in SLE T cells is not due to stability of the mRNA. These data are consistent with the postulate that estradiol stimulates CD154 transcription in SLE T cells.


Subject(s)
CD40 Ligand/genetics , Estradiol/pharmacology , Lupus Erythematosus, Systemic/physiopathology , RNA Stability/drug effects , T-Lymphocytes/physiology , Adult , Cells, Cultured , Estradiol/physiology , Female , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Middle Aged , RNA Stability/immunology , RNA, Messenger/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
4.
J Endocrinol ; 184(1): 119-27, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15642789

ABSTRACT

Differentiation of uterine stromal cells is critical for the establishment of pregnancy. This study had two purposes: (i) to validate the use of the UIII rat uterine stromal cell model for investigating mechanisms underlying decidual cell differentiation, and (ii) to use this cell model to identify a molecular switch for cellular entry into the decidual cell differentiation pathway. Quiescent rat uterine stromal cells were transfected with a 500 bp segment of the decidual prolactin-related protein (dPRP) promoter ligated to a luciferase reporter gene. Cells were incubated in low-serum medium, or in low-serum medium containing progesterone (1 microM), estradiol 17-beta (10 nM), cholera toxin (10 ng/ml) and interleukin-11 (10 ng/ml). Protein extracts were collected 48 h later and luciferase was measured in the cellular lysates. Cholera toxin and interleukin-11 stimulated luciferase expression (P< 0.05) and addition of sex steroids further increased (P< 0.05) dPRP promoter activity. Stromal cells did not proliferate (P< 0.05) under differentiation conditions. Deletion analysis of the dPRP promoter revealed maximal luciferase expression between -250 and -500 bp relative to the transcription start site. Comparison of cyclin E/Cdk2 activity between proliferating and differentiating cells showed a 3-fold increase (P< 0.05) at 12 h in differentiating cells. The results suggest that cyclin E/Cdk2 serves as a molecular switch for uterine stromal cell entry into the decidual cell differentiation pathway.


Subject(s)
Prolactin/analogs & derivatives , Prolactin/genetics , Stromal Cells/cytology , Uterus/cytology , Animals , Cell Differentiation , Cell Division , Cell Line , Cholera Toxin/pharmacology , Cyclin E/genetics , Cyclin E/metabolism , Estradiol/pharmacology , Female , Genes, Switch , Interleukin-11/pharmacology , Luciferases/analysis , Luciferases/genetics , Progesterone/pharmacology , Prolactin/metabolism , Promoter Regions, Genetic , Rats , Stimulation, Chemical , Stromal Cells/metabolism , Transfection/methods , Uterus/metabolism
5.
Placenta ; 22(8-9): 735-41, 2001.
Article in English | MEDLINE | ID: mdl-11597194

ABSTRACT

The H beta 58 gene, whose disruption in mice causes reabsorption of the embryo at 9.5 days post-conception, is believed to be essential for development of the placenta. Although the H beta 58 gene is well conserved in some Amniota, nothing is known about its presence in reptiles, some species of which have developed a chorioallantoic placenta. In this work, we investigated the expression of H beta 58 mRNA and protein in the three-toed skink, Chalcides chalcides. H beta 58 protein expression was found in the uterine epithelium beginning from the peri-ovulatory stage. However, it increased strongly at the moment of placental formation, when a high level of expression of mRNA and protein was also observed in the extra-embryonic membranes. The expression of H beta 58 mRNA and protein was maintained, although to a lesser degree, in the placenta during late pregnancy. It was also present in the early embryo. Finally, cloning and sequencing of a gene fragment revealed strong homology of the reptile gene with that of mammals. The high degree of conservation of the gene in amniote vertebrates and its presence in a viviparous squamate reptile (as in mammals) indicates an important role of this gene in the chorioallantoic placenta formation and development.


Subject(s)
Carrier Proteins/genetics , Placenta/physiology , Reptiles/genetics , Reptilian Proteins/genetics , Vesicular Transport Proteins , Allantois/chemistry , Animals , Base Sequence , Carrier Proteins/analysis , Chorion/chemistry , Cloning, Molecular , Epithelium/chemistry , Female , Gene Expression , Molecular Sequence Data , Ovulation , Pregnancy , RNA, Messenger/analysis , Rats , Reptilian Proteins/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Uterus/chemistry
6.
Int Immunopharmacol ; 1(6): 1009-24, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11407298

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease that occurs primarily in women (9:1 compared to men). Estrogen is a female sex hormone that acts on target cells through specific receptor proteins and alters the rate of transcription of target genes. Experiments in our laboratory have shown that calcineurin steady-state mRNA levels and phosphatase activity increase when estrogen is cultured with SLE T cells. This estrogen-dependent increase is dose-dependent, hormone-specific and temporally regulated. Estrogen receptor antagonism by ICI 182,780 inhibits the increase in calcineurin mRNA and phosphatase activity, while cycloheximide has no effect suggesting that new protein synthesis is not required. Reverse transcription and polymerase chain amplification indicate that estrogen receptor-alpha and estrogen-beta are expressed in human T cells. However, calcineurin does not respond to estrogen stimulation in T cells from normal females, males and lupus males. Taken together, these results indicate a differential function of the estrogen receptor in women with lupus. A model is proposed that suggests estrogen, acting through the estrogen receptor, enhances T cell activation in women with lupus resulting in amplified T-B cells interactions, B cell activation and autoantibody production.


Subject(s)
Autoimmunity/physiology , Estrogens/pharmacology , Lupus Erythematosus, Systemic/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Calcineurin/biosynthesis , Calcineurin/genetics , Female , Humans , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sex Characteristics
7.
Lupus ; 10(2): 116-22, 2001.
Article in English | MEDLINE | ID: mdl-11237123

ABSTRACT

We have shown that estrogen receptor (ERalpha, ERbeta) transcripts are expressed in SLE and normal T cells. In this study, T cell nuclear extracts from female lupus patients and normal donors were tested for biologically active ER proteins capable of binding to the human estrogen response element (hERE) by electrophoretic mobility shift assays. When peripheral blood T cells were stimulated with 17beta-estradiol (E2), PMA and ionomycin, two major retarded bands in T cell nuclear extracts exhibited a migration pattern similar to slow migrating protein-ERE complexes in human breast cancer cell extracts. T cells cultured only with E2 did not have these complexes. The formation of the complexes was inhibited by competition with the hERE cold oligonucleotide and partially with anti-ERalpha antibodies. There was no notable difference in the migration pattern of ERE-binding proteins between the SLE and normal T cell extracts. Together, these results suggest that activated human T cells, whether lupus-derived or normal-derived, contain biologically active ERalpha proteins. Other factors may be responsible for differential sensitivity of lupus T cells to estrogen.


Subject(s)
Estrogens/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Estrogen/immunology , T-Lymphocytes/immunology , Adult , Estrogens/metabolism , Female , Humans , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Middle Aged , Receptors, Estrogen/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism
8.
J Rheumatol ; 28(12): 2644-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11764210

ABSTRACT

OBJECTIVE: To examine the in vitro effects of estrogen on CD40 ligand (CD40L) expression in peripheral blood T cells isolated from patients with systemic lupus erythematosus (SLE) and normal controls. METHODS: T cells from female patients with SLE and controls were cultured in serum-free medium without and with 2-fluoroestradiol. Some T cells were activated by further culture on anti-CD3 coated plates. Calcineurin was activated in some T cells by culture in ionomycin. Cell surface CD40L was quantitated by FACS analysis. mRNA expression was measured using semiquantitative PCR. RESULTS: Lupus T cells cultured in medium containing 2-fluoroestradiol showed a significant (p = 0.04) increase in the amount of CD40L on the cell surface, but not in the number of positive cells, compared to the same T cells cultured without estradiol. Estradiol did not significantly change CD40L expression on the surface of T cells from normal women. In addition, the difference in cell surface CD40L between T cells cultured without and with estradiol was significantly greater (p = 0.048) on SLE than on normal T cells. Culture of SLE T cells in medium containing 2-fluoroestradiol followed by T cell receptor (TCR) activation for 2 h using anti-CD3 resulted in a significant (p = 0.04) estrogen dependent increase in CD40L mRNA. The estrogen dependent increases in SLE T cell CD40L mRNA and cell surface protein were blocked by the estrogen receptor antagonist ICI 182,780. SLE and normal T cells pretreated with estradiol and cultured with ionomycin for 2 h to activate calcineurin showed no significant differences in CD40L mRNA. CONCLUSION: These results suggest that estradiol, working through the estrogen receptor, stimulates the expression of CD40L in unstimulated and activated SLE T cells. Estradiol effects may be exerted on multiple regulatory steps that control CD40L expression. The estrogen dependent increase in CD40L expression could hyperstimulate SLE T cells and thereby contribute to the pathogenesis of SLE.


Subject(s)
CD40 Antigens/biosynthesis , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/pharmacology , Lupus Erythematosus, Systemic/blood , T-Lymphocytes/drug effects , Adult , CD3 Complex/immunology , CD40 Antigens/genetics , Cells, Cultured , Estrogen Antagonists/pharmacology , Female , Flow Cytometry , Fulvestrant , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Ligands , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
9.
Biol Reprod ; 63(6): 1735-46, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090444

ABSTRACT

Cloning and sequencing of a cDNA amplified by RNA fingerprinting at the implantation site of pregnant rats revealed 80% similarity with H beta 58, previously shown to be essential for formation of the chorioallantoic placenta in the mouse. H beta 58 mRNA was detected in the endometrium of hormonally sensitized rats stimulated to undergo decidualization and in the contralateral uterine horns lacking a decidual stimulus, indicating that uterine expression of H beta 58 mRNA did not require decidualization or the presence of a blastocyst. Immunodetection in the early postimplantation uterus (Days 6-8 of pregnancy) showed H beta 58 localized in the luminal and glandular epithelia and some stromal cells. Decidual cells at Day 6 of pregnancy expressed H beta 58, and by Day 9 of pregnancy, the protein localized throughout the maternal decidua. The temporal and spatial distribution of H beta 58 in the developing chorioallantoic placenta was assessed at Days 10, 12, and 14 of pregnancy. Immunoreactive H beta 58 localized to erythroid cells within the developing fetal vasculature of the chorioallantoic primordia at Day 10 of pregnancy. By Day 12, the fetal vasculature extended into the placental labyrinth, and the erythroid stem cells continued to strongly express H beta 58. At Day 14 of pregnancy, immunoreactivity became evident in the trophoblast giant cells and syncytiotrophoblast of the fetal placenta. As the chorioallantoic placenta matured (Day 18), H beta 58 mRNA was 3.6-fold higher in the labyrinth compared with the junctional region. Stable cell lines (HRP/LRP) isolated from the rat labyrinthine placenta expressed H beta 58 mRNA and protein. The expression pattern of H beta maternal and fetal placental tissues and its early expression in fetal erythroid stem cells during formation and maturation of the chorioallantoic placenta suggest that H beta 58 plays key roles in the regulatory networks that control hematopoietic development and placentation.


Subject(s)
Allantoin/metabolism , Carrier Proteins/biosynthesis , Chorion/metabolism , Gene Expression Regulation, Developmental/physiology , Placenta/metabolism , Vesicular Transport Proteins , Animals , Antibody Specificity , Blotting, Northern , Carrier Proteins/genetics , Cells, Cultured , Chorion/growth & development , DNA Fingerprinting , Embryo Implantation/physiology , Erythroid Precursor Cells/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/physiology , Immunohistochemistry , Male , Nuclease Protection Assays , Placentation , Pregnancy , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Uterus/metabolism , Uterus/ultrastructure
10.
Clin Immunol ; 95(2): 124-34, 2000 May.
Article in English | MEDLINE | ID: mdl-10779406

ABSTRACT

Previous experiments in our laboratory indicated that calcineurin expression and PP2B phosphatase activity increased when estrogen was cultured with SLE T cells but not with T cells from normal women. In this report we extended our findings to show that estrogen receptor (ER) antagonism by ICI 182,780 inhibited the estrogen-dependent increase in calcineurin mRNA and phosphatase PP2B activity indicating that estrogen action was mediated through the ER. Inhibition of de novo protein synthesis with cycloheximide suggested that the estrogen-dependent increase in T cell calcineurin mRNA was a direct effect of the ER and new protein synthesis was not required. Estrogen increased calcineurin mRNA in systemic lupus erythematosus (SLE) T cells at 6 h after the start of culture correlating with increased phosphatase activity at this same time. Phosphatase activity increased significantly (P < 0.02) in lupus T cells cultured for 8 h in estradiol-containing medium. Reverse transcription and polymerase chain amplification revealed that ER-beta and ER-alpha were expressed in female and male T cells from SLE patients and normal controls. However, calcineurin steady-state mRNA levels were unaffected by estradiol in cultured T cells from male SLE patients and normal male and female controls. These data indicate that estrogen, bound to the ER, evokes a direct increase in calcineurin expression in T cells from female lupus patients. This gender-specific response suggests that ER function is altered in women with the female predominant autoimmune disease, SLE.


Subject(s)
Calcineurin/genetics , Estrogens/pharmacology , Lupus Erythematosus, Systemic/pathology , Adolescent , Adult , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Prejudice , RNA, Messenger/metabolism , Receptors, Estrogen/physiology , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
11.
Endocrinology ; 141(2): 637-48, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650945

ABSTRACT

Understanding of cell cycle regulation in hormonally responsive cells lags behind studies in other systems because few models have been available to identify the role of steroid hormones and their receptors in this process. This study investigates progesterone-dependent effects on the progression of normal uterine stromal cells through early G1 phase of the cell cycle. Quiescent rat uterine stromal cells were stimulated to reenter the cell cycle by adding serum-free medium containing medroxyprogesterone acetate (MPA) and basic fibroblast growth factor (FGF). [3H]thymidine incorporation increased significantly (P = 0.025) in cells stimulated with both FGF alone and MPA plus FGF compared with the control cells. Moreover, cells stimulated with MPA plus FGF incorporated significantly more (P = 0.01) [3H]thymidine than cells treated with FGF alone, suggesting requisite interactions between progesterone and FGF for stromal cell entry into S phase. Flow cytometric analysis of stimulated stromal cells showed FGF alone and MPA plus FGF increased significantly (P = 0.002) the percentage of cells in S phase at 12 h. Incorporation of bromodeoxyuridine into stromal cell nuclei indicated that FGF alone and MPA plus FGF increased the percentage of cells entering S phase at 18 and 24 h compared with the control cells. In addition, MPA plus FGF increased significantly (P = 0.001) the number of cells entering S phase at 24 h compared with FGF alone and sustained S phase entry compared with FGF alone, MPA alone, or the control cells. Stromal cells inhibited from G1 reentry by inhibition of mitosis showed accelerated entry into S phase in response to MPA plus FGF compared with FGF alone. Cyclin D1 messenger RNA increased in stromal cells treated with MPA plus FGF at 9, 12, and 15 h. Addition of RU 486 to cells stimulated with MPA plus FGF for 9 h reduced cyclin D1 messenger RNA accumulation by 40%. Western blot analysis of cyclin D1 immunoprecipitates indicated complex formation with both cyclin-dependent kinase 4 (Cdk4) and cyclin dependent kinase 6 (Cdk6). Cyclin D1-Cdk complexes and kinase activity correlated temporally with increased cyclin D1 expression in cells cultured with MPA plus FGF. Taken together, these results show that progesterone-FGF interactions increase cyclin D1 expression, correlating with accelerated stromal cell entry into S phase compared with cells treated with FGF alone. Moreover, progesterone plus FGF sustains the timing of stimulation for transit of uterine stromal cells through G1 into S phase compared with FGF alone.


Subject(s)
Cell Cycle/physiology , Fibroblast Growth Factor 2/pharmacology , Medroxyprogesterone Acetate/pharmacology , Progesterone/physiology , Uterus/cytology , Animals , Cell Cycle/drug effects , Cells, Cultured , Cyclin D1/genetics , DNA/biosynthesis , Female , Flow Cytometry , G1 Phase , Gene Expression Regulation/drug effects , Kinetics , Ovariectomy , Progesterone/pharmacology , Rats , Rats, Sprague-Dawley , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/physiology , Thymidine/metabolism , Transcription, Genetic/drug effects
12.
Clin Immunol Immunopathol ; 89(2): 171-80, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9787119

ABSTRACT

Systemic lupus erythematosus (SLE) predominantly affects women (9:1 compared to men) of childbearing age and often decreases its intensity in postmenopausal women, suggesting that sex hormones play a role in its pathogenesis. Comparison of steady-state levels of calcineurin mRNA using RNase protection assays revealed increased calcineurin expression in response to estradiol in cultured T cells from nine female lupus patients. Calcineurin mRNA levels did not increase significantly in T cells from eight age-matched normal control female volunteers. Estrogen-dependent calcineurin mRNA increased in a dose-dependent fashion, while progesterone and dexamethasone did not increase calcineurin mRNA in patient cells. Lupus T cell calcineurin mRNA increased in response to estradiol at 6 h but not at 3 h. Calcineurin phosphatase activity increased in lupus T cell extracts after incubation of cells with estradiol, while phosphatase activity in normal T cells was unaffected by estrogen. Calcineurin expression in T cells from patients with vasculitis and rheumatoid arthritis taking medications similar to those taken by the lupus patients was unaffected by estradiol. This study provides the first evidence for a molecular marker of estrogen action in lupus patients and suggests that estrogen-dependent changes in lupus T cell calcineurin could alter proinflammatory cytokine gene regulation and T-B cell interactions.


Subject(s)
Autoimmune Diseases/physiopathology , Adult , Calcineurin/genetics , Calcineurin/pharmacology , Cells, Cultured , Culture Media , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogens/pharmacology , Female , Gene Expression/drug effects , Humans , Lupus Erythematosus, Systemic/genetics , Lymphocyte Activation , Male , RNA, Messenger/metabolism , Sex Characteristics , Sex Ratio , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Time Factors
14.
J Rheumatol ; 25(7): 1305-12, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9676761

ABSTRACT

OBJECTIVE: To identify and characterize estrogen receptor (ER) transcripts expressed in immune cells of patients with systemic lupus erythematosus (SLE) and healthy donors. METHODS: Peripheral blood monocytes and T cells were prepared from patients with SLE (n = 6) and healthy donors (n = 8). T cells were separated into CD4 and CD8. Some monocytes and T cells were stimulated with estradiol, PMA, and ionomycin. Epstein-Barr virus-transformed B cell lines (n = 7) and B cell hybridomas (n = 2) established from patients with SLE and a healthy individual were used as a B cell source. These cells were examined for ER mRNA by reverse transcription nested polymerase chain reaction. Amplified cDNA were sequenced by standard methods. RESULTS: In all cells tested, ER mRNA was expressed without prior in vitro stimulation. Partial sequences from exons 1-8 were nearly identical to the published sequence of the human ER mRNA. There were no notable differences in the ER transcripts between patients and healthy controls. Variant receptor transcripts lacking exon 5 or exon 7, which encodes the hormone binding domain, were identified in the majority of the cells. Precise deletion of the exons suggests that they are alternatively spliced transcripts. Whether the detected transcripts are translated into functional receptor proteins remains to be determined. In vitro stimulation did not affect ER mRNA expression. The presence of variants did not correlate with disease activity or medication. CONCLUSION: Monocytes, T cells, and B cells in patients express transcripts of the normal wild type ER and the hormone binding domain variants in vivo.


Subject(s)
B-Lymphocytes/metabolism , Lupus Erythematosus, Systemic/metabolism , Monocytes/metabolism , Receptors, Estrogen/metabolism , T-Lymphocytes/metabolism , Adult , B-Lymphocytes/drug effects , Cell Line, Transformed , Estradiol/pharmacology , Female , Humans , Ionomycin/pharmacology , Lupus Erythematosus, Systemic/genetics , Monocytes/drug effects , Polymerase Chain Reaction , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Sequence Analysis , T-Lymphocytes/drug effects , Tetradecanoylphorbol Acetate/pharmacology
15.
J Endocrinol ; 154(1): 75-84, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9246940

ABSTRACT

Cell proliferation and differentiation in the rodent uterus are probably controlled by the interaction of female sex steroids with polypeptide growth factors. Uterine basic fibroblast growth factor (bFGF) mRNA was measured by RNase protection during the time (days 2-4) of endometrial cell proliferation in the pregnant rat. bFGF transcripts were detected at each of the 3 days of pregnancy examined. To investigate the influence of oestrogen and progesterone on bFGF mRNA accumulation, ovariectomized rats were treated with oestradiol for 48 h followed by a single injection of oestradiol, progesterone, the two steroids co-injected or oil vehicle alone. Uterine RNA was collected 6 h after the last hormone injection. Steroid treatments increased steady-state uterine bFGF mRNA compared with vehicle control animals as measured by RNase protection. Northern blot analysis of c-fos and c-jun mRNAs from these same treatment groups revealed increased protooncogene expression in the uterus of hormone treated rats compared with the control animals. Temporal analysis of bFGF mRNA in ovariectomized rats at 1, 3 and 6 h after acute oestrogen and oestrogen-progesterone co-administration showed a dual pattern of transcript accumulation. Both hormone treatments increased bFGF mRNA within 1 h compared with vehicle injected rats. Co-administration of the two hormones, however, repressed bFGF mRNA accumulation relative to oestrogen at 3 and 6 h. Together, these studies provide evidence that bFGF control of uterine cell proliferation in pregnant rats can occur from newly synthesized bFGF. Moreover, the results suggest that progesterone is a potent stimulator of bFGF expression in the uterus.


Subject(s)
Estradiol/pharmacology , Fibroblast Growth Factor 2/metabolism , Progesterone/pharmacology , RNA, Messenger/metabolism , Uterus/drug effects , Animals , Blotting, Northern , Female , Fibroblast Growth Factor 2/genetics , Ovariectomy , Pregnancy , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-jun/analysis , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Uterus/metabolism
16.
Biol Reprod ; 55(6): 1333-42, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8949891

ABSTRACT

Uterine stromal cells undergo mitosis and differentiate into the decidua just prior to the expected time of implantation in humans and rodents. We have utilized a culture system that will be suitable for study of the molecular mechanisms regulating stromal cell proliferation. Stromal cells were isolated from the uteri of ovariectomized rats and were cultured in chemically defined medium. Cultured cells express the mesenchymal markers vimentin and desmin. They do not express the epithelial marker cytokeratin. Serum-starved stromal cells were stimulated to proliferate in a time frame consistent with the cell cycle through addition of a panel of growth factors (basic fibroblast growth factor [bFGF], epidermal growth factor, platelet-derived growth factor, transforming growth factor alpha, insulin-like growth factor I) and hormones to the culture medium. None of the growth factors tested significantly stimulated proliferation in the absence of progesterone. Furthermore, progesterone was the only steroid of those tested that stimulated mitosis in the presence of growth factors. Stromal cell proliferation in response to progesterone and bFGF was dose dependent and saturable. Addition of the progesterone receptor antagonist mifepristone (RU486) and an inhibitor of tyrosine kinase receptor activation (suramin) abolished stromal cell mitosis. Progesterone receptors and fibroblast growth factor receptor 1 (FGFR1) were identified by immunoblot analysis in proliferating stromal cells. Taken together, these results show that cultured stromal cells maintain progesterone-dependent cell cycle control that is mediated via progesterone receptors. Moreover, the data indicate that bFGF control of stromal cell proliferation is modulated via a specific isoform of FGFR1 containing the three-loop immunoglobulin-like domain.


Subject(s)
Cell Division/drug effects , Growth Substances/pharmacology , Progesterone/pharmacology , Stromal Cells/cytology , Uterus/cytology , Animals , Cells, Cultured , Culture Media , Desmin/analysis , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Growth Substances/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Platelet-Derived Growth Factor/pharmacology , Progesterone/analysis , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/analysis , Stromal Cells/chemistry , Transforming Growth Factor alpha/pharmacology , Vimentin/analysis
17.
Endocrinology ; 136(7): 3137-45, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7789341

ABSTRACT

Recent studies suggest that steroid effects on uterine cell proliferation may be moderated by polypeptide growth factors. We now provide evidence that high affinity fibroblast growth factor (FGF) receptors are present temporally and spatially in the pregnant rat uterus (days 4-6) to support the idea that basic FGF action occurs via binding to its high affinity FGF receptor 1 (FGFR1). Reverse transcription-polymerase chain amplification indicates that both the full-length transcript and an alternatively spliced messenger RNA are present in the uterus. Western immunoblot analysis confirms that rat uterine membrane proteins contain two receptor isoforms, and these receptors bind basic FGF with high affinity and specificity. Immunolocalization of FGFR1 revealed receptor-positive cells in both the uterine stroma and epithelia on days 4-6 of pregnancy. However, the receptor was differentially localized in the disparate cell types. The nuclei of stromal cells were positive for FGFR1, whereas epithelial cell nuclei were negative. Together, these results suggest that FGF signal transduction in uterine stromal cells is mediated by activation of FGFR1.


Subject(s)
Alternative Splicing , Pregnancy, Animal , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/genetics , Uterus/metabolism , Animals , Base Sequence , Blotting, Western , Cell Membrane/chemistry , Cell Nucleus/chemistry , Epithelium/chemistry , Epithelium/ultrastructure , Female , Fibroblast Growth Factor 2/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Pregnancy , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/analysis , Stromal Cells/chemistry , Stromal Cells/ultrastructure , Uterus/chemistry
18.
Microsc Res Tech ; 27(2): 125-33, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-8123905

ABSTRACT

Electron and fluorescence microscopic techniques have been used in a complementary fashion to study the patterns of follicle cell-oocyte interactions within cumulus-oocyte-complexes of various mammals. The principal findings are: (1) two distinct types of transzonal processes exist that are distinguishable on the basis of cytoskeletal composition; (2) in some of the species examined (pig, goat, primate), corkscrew-shaped processes rich in tubulin, traverse the zona pellucida and are invaginated into the oocyte cortex; (3) actin-rich processes either ramify as a network at the outer surface of the zona pellucida or penetrate the zona and make contact with the oolemma in a species specific manner. These results are discussed with respect both to the need to employ complementary optical methods in assessing connectivity patterns within COC and to the possible role that extracellular matrix-cell interactions play in the homeostatic control of oocyte growth and maturation.


Subject(s)
Cell Communication , Oocytes/cytology , Ovarian Follicle/cytology , Animals , Cells, Cultured , Female , Guinea Pigs , Horses , Macaca mulatta , Microscopy, Electron , Microscopy, Fluorescence , Oocytes/physiology , Oocytes/ultrastructure , Rats , Swine
19.
J Endocrinol ; 140(2): 239-49, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8169558

ABSTRACT

Recent studies suggest that hormonal control of uterine cell proliferation may be moderated by polypeptide growth factors. It remains to be determined, however, whether growth factors cause or are the consequence of hormone action. Basic fibroblast growth factor (bFGF) has been shown to influence cell proliferation and differentiation of a variety of mesoderm-derived cells. To elucidate the regulatory mechanisms controlling stromal cell proliferation and differentiation required for embryo implantation further, immunohistochemical localization of the progesterone receptor and bFGF have been studied. The cell-specific distribution of these proteins was determined in the rat uterus during early pregnancy and after injection of the progesterone receptor antagonist mifepristone (RU 486) at days 1 and 2 post coitum (p.c.) to block implantation. Cell division was restricted to luminal and glandular epithelial cells in pregnant and RU 486-treated rats at day 3 p.c. At day 4 of pregnancy, cell proliferation switched from the epithelia to the stroma in pregnant rats, but after RU 486 treatment division of stromal cells was inhibited significantly (P < 0.05). Progesterone receptor distribution was altered and bFGF was absent in RU 486-blocked stromal cells. Expression of bFGF in luminal and glandular epithelial cells, however, was insensitive to the effects of progesterone receptor antagonism. bFGF content was stimulated in the luminal epithelium and in decidual cells by the implanting embryo. These results indicate that repression of progesterone receptor function in early pregnancy results in a cell-specific loss of bFGF from stromal cells and inhibition of their proliferation. The results further suggest that the regulation of endometrial cell bFGF content is modulated at the site of implantation by the embryo.


Subject(s)
Embryo Implantation/physiology , Fibroblast Growth Factor 2/metabolism , Pregnancy, Animal/metabolism , Receptors, Progesterone/metabolism , Uterus/metabolism , Animals , Cell Differentiation/physiology , Cell Division/physiology , Female , Immunohistochemistry , Mifepristone/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/drug effects , Uterus/cytology
20.
Biol Reprod ; 49(4): 653-65, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8218629

ABSTRACT

Cellular proliferation and differentiation are critical components of uterine remodeling prior to embryonic implantation. Recent studies have shown that the ovarian hormones, estrogen and progesterone, modulate these cellular events through the production of growth factors. Basic fibroblast growth factor (bFGF) has been implicated in the control of cell proliferation, differentiation, and embryonic development. To clarify its role in uterine remodeling, the cellular distribution of bFGF was examined immunohistochemically in the rat uterus during early pregnancy (Days 2-6). Basic FGF localized intracellularly in stromal and epithelial cells and within the extracellular matrix at Days 2 and 3. It was distinctly evident at the apical surface of epithelial cells at Days 4 and 5 of pregnancy. Concurrent with this apical localization, bFGF was present in the uterine luminal fluid, suggesting release of this growth factor from epithelial cells. Embryonic implantation was accompanied by increased intracellular bFGF content in luminal epithelial and decidual cells. However, similar cells outside of the implantation site and in the artificially decidualized uterus did not express analogous bFGF levels, indicating that a unique signal from the embryo triggers bFGF expression. Changes in the cell-specific distribution of bFGF imply a multifunctional role for this growth factor in uterine cell proliferation, differentiation, and embryonic implantation. In addition, the apical release of bFGF from epithelial cells indicates utilization of a novel secretory pathway for bFGF export during early pregnancy.


Subject(s)
Embryo Implantation/physiology , Fibroblast Growth Factor 2/metabolism , Uterus/metabolism , Animals , Cell Differentiation , Cell Division , Decidua/metabolism , Epithelial Cells , Epithelium/metabolism , Female , Immunoenzyme Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution , Uterus/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...