Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Br J Cancer ; 102(8): 1276-83, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20354527

ABSTRACT

BACKGROUND: Ovarian cancer is the most lethal gynaecological malignancy. Although ovarian cancer patients often respond initially to chemotherapy, they usually develop chemoresistance. We hypothesised that a small portion of ovarian cancer cells have stem-like cell properties that contribute to tumourigenesis and drug resistance. METHODS: Flow cytometry and Hoechst 33342 efflux isolated side-population (SP) cells from ascites derived from ovarian cancer patients and from mice inoculated with human ovarian cancer cell lines. The SP cells were examined for stem cell markers OCT4, NANOG, STELLAR, and ABCG2/BCRP1 by immunocytochemistry and RT-PCR. The SP cells and non-SP cells were studied for tumourigenesis and chemoresistance in vitro and in vivo. RESULTS: The SP cells expressed ABCG2/BCRP1, OCT4, STELLAR, and NANOG, detected by immunocytochemistry and RT-PCR. ABCG2/BCRP1 expression was higher in SP than in non-SP cells. Xenogeneic mice inoculated with SP cells yielded more tumours than did mice inoculated with non-SP cells. In parallel, SP cell culture resulted in extensive cell proliferation, which was markedly more than in non-SP cells. SP cells resisted chemotherapy compared with non-SP cells, both in vivo and in vitro. CONCLUSION: Ovarian cancer SP cells are tumourigenic and chemoresistant. ABCG2/BCRP1 has an important role in chemoresistance, which has implications for new therapeutic approaches.


Subject(s)
Drug Resistance, Neoplasm , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Ascitic Fluid/pathology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Transplantation
2.
Mol Hum Reprod ; 11(4): 253-60, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15734895

ABSTRACT

Angiopoietin (Ang)-2, the natural antagonist of the Ang1/Tie2 receptor is a complex regulator of blood vessel plasticity that plays a pivotal role in both vessel sprouting [in the presence of vascular endothelial growth factor (VEGF)-A] and vessel regression (in the absence of VEGF-A). Based on the spatial and temporal expression of Ang2 throughout human gestation, we recently suggested that the Ang2 may play a pivotal role in placental angiogenesis. Further, to examine this tenet we have developed a novel murine model system in which in utero Ang2 gene delivery via a non-replicating adenoviral expression vector has the potential to manipulate the blood vessel phenotype in vivo during pregnancy. Ang2 overexpression selectively and rapidly remodels the labyrinth perivascular extracellular matrix, subsequently promoting plasticity of the maternal and fetal vessels, which appear honeycombed due to a 2-fold increase in blood vessel luminal area. High levels of Ang2 impair endothelial cell adhesiveness, leading to vascular leakiness with perivascular oedema, which increases placental weight. These observations suggest that the Ang2 overexpression may play a key role in placental vascular remodelling. Furthermore, we suggest a novel new model to study the pathobiology of placental vascularization and the effect of placental blood vessels on fetal phenotype.


Subject(s)
Angiopoietin-2/physiology , Neovascularization, Physiologic , Placenta/blood supply , Angiopoietin-2/analysis , Angiopoietin-2/genetics , Animals , Blood Vessels/cytology , Blood Vessels/growth & development , Endothelium, Vascular/chemistry , Extracellular Matrix/chemistry , Female , Genetic Vectors , Humans , Mice , Models, Animal , Neovascularization, Physiologic/physiology , Phenotype , Placenta/chemistry , Placentation , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection
4.
J Clin Endocrinol Metab ; 86(9): 4512-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549701

ABSTRACT

Hormones produced by the fetal adrenal regulate fetal growth, steroidogenic activity, and intrauterine homeostasis, which are essential for the maintenance of pregnancy and the preparation of the fetus for extrauterine life. There is a functional interaction between CRH and the fetal adrenal, as CRH increases dehydroepiandrosterone sulfate production in cultured fetal adrenal cells. Moreover, in a rodent model administration of orexin A induced corticosterone production. To examine this relationship in more detail we measured the expression of the different subtypes of CRH and orexin receptors and their specific coupling to G protein alpha-subunits upon activation with CRH and orexin A, respectively. Using RT-PCR and fluorescent in situ hybridization analysis, we demonstrated the presence of CRH receptors 1alpha and 2alpha, and orexin type 2 receptor mRNA. None of the other CRH receptor variants or orexin type 1 receptor were detected. Immunofluorescent analysis and Western blotting confirmed the protein expression of both receptors, which also bind fluo-CRH and fluo-orexin with high affinity. Immunoblotting analysis confirmed the expression of prepro-orexin and orexin A in fetal adrenals. Using photoaffinity labeling, we determined which G proteins are coupled to the CRH and orexin receptors in fetal adrenals when challenged with CRH or orexin. Treatment of fetal adrenal membranes with CRH (100 nM) increased the labeling of G(o) and, to a lesser extent, G(s), but not G(i) and G(q), whereas treatment with orexin A (100 nM) increased the labeling of G(s) and G(i), but not G(o) and G(q). These findings provide new insights into the components of the signal transduction machinery in human fetal adrenals and demonstrate for the first time the presence of functional orexin receptors outside of the CNS in humans.


Subject(s)
Adrenal Glands/embryology , Adrenal Glands/metabolism , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Neuropeptide/biosynthesis , Adult , Blotting, Western , Female , Fluorescent Antibody Technique , Fluorescent Dyes , Humans , In Situ Hybridization, Fluorescence , Orexin Receptors , Photoaffinity Labels , Pregnancy , Receptors, G-Protein-Coupled , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
5.
Biochem J ; 359(Pt 1): 219-26, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11563986

ABSTRACT

Vascular endothelial growth factor (VEGF) is a mitogen in physiological and pathological angiogenesis. Understanding the expression of different VEGF isoforms might be important for distinguishing angiogenesis in tissue development, vascular remodelling and tumour formation. We examined its expression and noted the presence of the isoforms VEGF(121) and VEGF(165) (121 and 165 residues long respectively) in fetal heart, lung, ovary, spleen, placenta and ovarian tumours. Unexpectedly, a 47 kDa species predominated in fetal intestine and muscle. The presumed initiation site in VEGF is an AUG codon (AUG(1039)), 1039 nt from its main transcriptional start site. AUG(1039) is preceded in the 5' untranslated region by an in-frame CUG at nt 499 (CUG(499)), which could produce the 47 kDa form with a 180-residue N-terminal extension. We therefore assessed whether CUG(499) functions as an initiator. CUG(499) initiation produced the 47 kDa VEGF(165) precursor, which was processed at two sites to yield VEGF and three N-terminal fragments. When CTG(499) was mutated to CGC, the precursor and N-terminal fragments were barely detectable. Although the precursor form was predominant in VEGF(165), both CUG(499) and AUG(1039) forms were found in VEGF(121). VEGF precursor induced neither the proliferation of human umbilical vein endothelial cells nor the expression of angiopoietin 2, which can be induced by, and act with, VEGF to induce tumour angiogenesis. The precursor also adheres to the extracellular matrix (ECM), suggesting that it might be a storage form for generating active VEGF in the cell or ECM. Alternate CUG(499) and AUG(1039) initiation and processing of the inactive precursor and its products might be important in regulating angiogenesis.


Subject(s)
Codon, Initiator/genetics , Endothelial Growth Factors/metabolism , Fetus/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Ovarian Neoplasms/metabolism , Ribosomes/metabolism , 5' Untranslated Regions/genetics , Animals , Blotting, Western , COS Cells , Cell Division , Chlorocebus aethiops , Endothelial Growth Factors/genetics , Extracellular Matrix , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Mutation/genetics , Ovarian Neoplasms/genetics , Pregnancy , Pregnancy Trimester, Second , Protein Biosynthesis , Protein Isoforms , Protein Precursors/genetics , Protein Precursors/metabolism , Sequence Deletion , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
J Natl Cancer Inst ; 93(10): 762-8, 2001 May 16.
Article in English | MEDLINE | ID: mdl-11353786

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) stimulates ovarian tumor growth at concentrations present in ascitic fluid. Vascular endothelial growth factor (VEGF) stimulates angiogenesis and plays a pivotal role in the formation of ovarian cancer-associated ascites. We examined whether LPA promotes ovarian tumor growth by increasing angiogenesis via VEGF. METHODS: VEGF expression was examined in a simian virus 40 T-antigen-immortalized ovarian surface epithelial cell line (IOSE-29) and in ovarian cancer cell lines (OVCAR-3, SKOV-3, and CAOV-3) treated with LPA. VEGF promoter activity was measured in OVCAR-3 cells after transfection or cotransfection with c-Fos and c-Jun, components of AP1 transcription factor, potential binding sites for which are present in the VEGF promoter. The expression of the LPA receptors Edg2 and Edg4 was also assessed. All statistical tests were two-sided. RESULTS: LPA treatment increased steady-state VEGF messenger RNA (mRNA) levels in OVCAR-3 cells in a time- and dose-dependent fashion and stimulated VEGF promoter activity without prolonging mRNA half-life in these cells, but LPA had little effect on IOSE-29 cells. Forced overexpression of c-Jun and c-Fos in OVCAR-3 cells stimulated VEGF promoter activity fourfold. LPA also elevated VEGF protein levels by 1.5-fold in SKOV-3 cells (P =.0148), 1.9-fold in CAOV-3 cells (P<.001), and threefold in OVCAR-3 cells (P<.0001). Both Edg2 and Edg4 were detected in ovarian cancer cells; however, only Edg2 was present in normal ovarian surface epithelial cells and IOSE-29 cells. CONCLUSIONS: LPA stimulates ovarian tumor growth, at least in part, via induction of VEGF expression through transcriptional activation. However, this LPA response is not evident in normal ovarian surface epithelial cells. Our data suggest that Edg4, but not Edg2, plays a role in LPA stimulation of ovarian tumor growth.


Subject(s)
Endothelial Growth Factors/biosynthesis , Lymphokines/biosynthesis , Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Receptors, G-Protein-Coupled , Antibiotics, Antineoplastic/pharmacology , Binding Sites , Blotting, Northern , Cell Line , Cell Line, Transformed , Culture Media, Conditioned/metabolism , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Gene Deletion , Humans , Luciferases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface/biosynthesis , Receptors, Lysophosphatidic Acid , Recombinant Fusion Proteins/metabolism , Time Factors , Transcription Factor AP-1/metabolism , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Maturitas ; 38(2): 205-10, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11306210

ABSTRACT

OBJECTIVES: Estrogen is often prescribed for symptoms and sequelae of ovarian estrogen loss after menopause. METHODS: To assess efficacy and acceptability of a new, highly soluble estrogen-calcium preparation, we formulated a water-soluble powdered combination of estrogen (0.625 mg estrone piperazine sulfate) and calcium (1 g, ions) as the highly soluble glycerophosphate salt (Estrosol). Effects of once-daily administration on bone mineral turnover of Estrosol dissolved in water (n = 11) was compared with 0.625 mg conjugated estrogens (Premarin) + 1 g calcium (Tums 500 Calcium Supplement) (n = 8). All women had had a previous hysterectomy, were between the ages 40 and 75, within 25% of ideal body weight, and had not taken hormonal preparations for at least 3 months. Assessment of bone mineral turnover was by monitoring N-telopeptides and bone specific alkaline phosphatase (BSAP) on 5 occasions: pretreatment and once during each of the 4 months of treatment. RESULTS: Mean N-telopeptide values decreased (p = .005) in both groups: Estrosol, 29.2% (40 --> 29 mmol bone collagen equivalents (BCE)/mmol creatinine), and Premarin(R) + calcium, 44.8% (33 --> 18 mmol). Mean BSAP values also decreased (p = 0.007) in both groups: Estrosol, 12.6% (12.06 --> 10.54 mg/l), Premarin(R) + calcium, 19.1% (11.57 --> 9.36 mg/l). The difference between groups for both N-telopeptides and BSAP was not significant, although sample size was small. Symptoms (hot flashes, vaginal dryness) improved similarly in both groups. Symptoms during treatment (breast or nipple tenderness, bloating) also were similar in both groups. Both preparations were well-tolerated. There were no changes in CBC, liver function tests, electrolytes or urinalyses in either group . CONCLUSIONS: This pilot study indicates that the combined, highly water-soluble preparation of estrogen and calcium is effective in reducing bone mineral turnover, acceptable and well-tolerated. Use of this single aqueous preparation may lead to better compliance than using two separate pills.


Subject(s)
Bone Density/drug effects , Calcium/pharmacology , Estrogens, Conjugated (USP)/pharmacology , Estrone/pharmacology , Hormone Replacement Therapy , Osteoporosis, Postmenopausal/prevention & control , Adult , Aged , Alkaline Phosphatase/blood , Calcium/administration & dosage , Chemistry, Pharmaceutical , Drug Administration Schedule , Estrogens, Conjugated (USP)/administration & dosage , Estrone/administration & dosage , Female , Humans , Middle Aged , Pilot Projects
9.
Am J Primatol ; 53(3): 123-30, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11253847

ABSTRACT

In humans, placental corticotropin-releasing hormone (CRH) production has been linked to the determination of gestational length, and a late gestational fall in CRH-binding protein (CRH-BP) has been linked to the onset of parturition. Expression of placental CRH mRNA is limited to primates, and only in man has a circulating CRH-BP been described. As the fall in CRH-BP in late gestation has been associated with parturition in humans, we sought to determine whether a CRH-BP circulated in the plasma of other primates. It is unclear whether maternal plasma CRH concentrations are elevated in New World monkeys and prosimians. We have therefore performed CRH plasma measurements in the blood of pregnant marmosets, in several species of lemur, and in pregnant and fetal rhesus monkeys as a positive control. Using gel chromatography, CRH-BP was detected in the human, gorilla, chimpanzee, orangutan, gibbon, macaque, squirrel monkey, and marmoset, but was absent in the mandrill, spider monkey, and lemur. CRH was detected in the plasma of pregnant marmosets and rhesus monkeys. CRH was also detected in the fetal rhesus monkey, but at lower concentrations than in maternal plasma. CRH immunoreactivity was not detectable in the plasma of pregnant lemurs or in extracts of lemur placenta. In conclusion, a circulating binding protein for CRH exists in all species of apes but occurs variably among New World and Old World monkeys and is absent in lemurs. The variable occurrence of the CRH-BP does not support a role for this protein in the mechanism of parturition in primates. Maternal CRH is elevated in the pregnant marmoset and rhesus, and may play a role in the pregnancy of New and Old World monkeys.


Subject(s)
Carrier Proteins/analysis , Pregnancy/physiology , Primates/physiology , Animals , Carrier Proteins/pharmacology , Chromatography, Gel , Corticotropin-Releasing Hormone/analysis , Corticotropin-Releasing Hormone/pharmacology , Female , Humans
10.
Am J Obstet Gynecol ; 183(4): 956-63, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11035346

ABSTRACT

OBJECTIVE: Magnetic resonance imaging enhanced with macromolecular contrast medium was used to monitor effects of angiogenesis inhibition on tumor microvascular permeability and ascites volume in an athymic rat model of human ovarian cancer. STUDY DESIGN: Groups of 6 athymic rats implanted intraperitoneally with SKOV-3, a human ovarian cancer cell line, were treated through a 14-day course with antibody to vascular endothelial growth factor or with saline solution for control animals. Dynamic magnetic resonance imaging was performed with a 92,000-d contrast agent, albumin-(gadolinium-diethylenetriaminepentaacetic acid)(30). Vascular permeability was estimated from dynamic enhancement data that were analyzed with a unidirectional 2-compartment kinetic model. RESULTS: Animals treated with vascular endothelial growth factor antibody accumulated significantly smaller volumes of peritoneal ascites (P <.05) and showed significantly lower magnetic resonance imaging-assayed tumor microvascular permeabilities (P <.05) than did control animals. CONCLUSION: Magnetic resonance imaging enhanced with a macromolecular contrast agent in an athymic rat model of human ovarian cancer treated with anti-vascular endothelial growth factor antibody can be used to measure a reduction in tumor microvascular permeability, corresponding to a reduction in ascites production.


Subject(s)
Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Magnetic Resonance Imaging , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/physiopathology , Animals , Antibodies/pharmacology , Ascites/prevention & control , Capillary Permeability , Endothelial Growth Factors/immunology , Female , Humans , Lymphokines/immunology , Neoplasm Transplantation , Peritoneal Diseases/prevention & control , Rats , Rats, Nude , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Fertil Steril ; 74(3): 429-38, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973633

ABSTRACT

OBJECTIVE: To review the current literature on the role of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) in ovarian physiology and pathology. DESIGN: A computerized search was conducted to identify relevant in vitro and in vivo studies published in English. MEDLINE, Current Contents, and the Index Medicus were searched for studies published before January 2000. RESULT(S): VEGF/VPF is an angiogenic factor and a potent mitogen for vascular endothelium. During reproductive life, VEGF/VPF plays a role in the cyclic growth of ovarian follicles and corpus luteum development and maintenance, mediating ovarian angiogenesis. VEGF/VPF expression and secretion are induced by both FSH and LH/hCG receptor-activated pathways. CONCLUSION(S): VEGF/VPF expression and production within the ovary are critical for normal reproductive function. Defects in angiogenesis may contribute to a variety of disorders including anovulation and infertility, pregnancy loss, ovarian hyperstimulation syndrome, and ovarian neoplasms.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Ovary/physiology , Animals , Female , Humans , MEDLINE , Ovary/blood supply , Ovary/pathology , Pregnancy , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Hum Reprod ; 15(9): 1889-97, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966981

ABSTRACT

Both Fas (APO-1, CD95), an apoptosis-inducing receptor, and its ligand, Fas ligand (FasL, CD95L), have been localized to the ovary. Granulosa cell apoptosis occurs in antral follicular atresia. In polycystic ovary syndrome (PCOS), antral follicles accumulate with some atretic features. The ovarian expression of Fas and FasL was examined in PCOS by immunohistochemistry and correlated with immunodetection of apoptotic cells. Fas immunostaining was present in pre-antral follicle oocytes, some primary and secondary pre-antral follicle granulosa cells, and both granulosa and theca of antral follicles. Thecal staining persisted with advancing atresia, while granulosa staining declined. In antral follicles, abundant Fas-positive cells co-localized with scattered nuclei immunopositive for apoptosis. Ovarian vascular myocytes were strongly Fas-immunopositive. FasL immunostaining was present in pre-antral follicles in oocytes and variably in granulosa. In antral follicles, granulosa and thecal FasL staining increased with advancing atresia. Normal control ovaries showed follicular Fas and FasL staining patterns similar to those in PCOS, but vascular staining was less prominent. In one healthy follicle, Fas immunostaining was seen in the oocyte and weakly in mural granulosa and theca interna. The results suggest that in PCOS, an alteration in Fas-mediated apoptosis, does not cause abnormal folliculogenesis, but may promote ovarian vascular remodelling.


Subject(s)
Apoptosis , Membrane Glycoproteins/analysis , Ovary/chemistry , Polycystic Ovary Syndrome/metabolism , fas Receptor/analysis , Adult , Corpus Luteum/chemistry , Fas Ligand Protein , Female , Granulosa Cells/chemistry , Humans , Immunohistochemistry , Ovarian Follicle/chemistry , Theca Cells/chemistry
13.
J Lipid Res ; 41(8): 1231-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946010

ABSTRACT

Apolipoprotein L is a newly recognized component of human plasma lipoproteins. Mainly associated with apoA-I-containing lipoproteins, it is a marker of distinct HDL subpopulations. In an effort to gain inference as to its as yet unknown function, we studied biological determinants of apoL levels in human plasma. The distribution of apoL in normal subjects is asymmetric, with marked skewing toward higher values. No difference was found in apoL concentrations between males and females, but we observed an elevation of apoL in primary hypercholesterolemia (10.1 vs. 8.5 microgram/mL in control), in endogenous hypertriglyceridemia (13.8 microgram/mL, P < 0.001), combined hyperlipidemia phenotype (18.7 g/mL, P < 0.0001), and in patients with type II diabetes (16.2 microgram/mL, P < 0.02) who were hyperlipidemic. Significant positive correlations were observed between apoL and the log of plasma triglycerides in normolipidemia (0.446, P < 0.0001), endogenous hypertriglyceridemia (0.435, P < 0.01), primary hypercholesterolemia (0.66, P < 0.02), combined hyperlipidemia (0.396, P < 0.04), hypo-alphalipoproteinemia (0.701, P < 0.005), and type II diabetes with hyperlipidemia (0.602, P < 0. 01). Apolipoprotein L levels were also correlated with total cholesterol in normolipidemia (0.257, P < 0.004), endogenous hypertriglyceridemia (0.446, P = 0.001), and non-insulin-dependent diabetes mellitus (NIDDM) (0.548, P < 0.02). No significant correlation was found between apoL and body mass index, age, sex, HDL-cholesterol or fasting glucose and glycohemoglobin levels. ApoL levels in plasma of patients with primary cholesteryl ester transfer protein deficiency significantly increased (7.1 +/- 0.5 vs. 5.47 +/- 0.27, P < 0.006).


Subject(s)
Apolipoproteins/blood , Cholesterol/blood , Diabetes Mellitus, Type 2/blood , Glycoproteins , Hyperlipidemias/blood , Lipoproteins, HDL/blood , Triglycerides/blood , Adult , Aged , Aged, 80 and over , Apolipoprotein L1 , Carrier Proteins/genetics , Cholesterol Ester Transfer Proteins , Female , Humans , Hypercholesterolemia/blood , Hypertriglyceridemia/blood , Male , Middle Aged , Mutation , Tangier Disease/blood
14.
Obstet Gynecol Surv ; 55(8): 511-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10945194

ABSTRACT

UNLABELLED: Components of the female reproductive system undergo a number of programmed angiogenic processes coupled with cyclic evolution and decline of ovarian, endometrial, and placental structures. The development of a new vascular network requires a remarkable degree of coordination between different cell types undergoing complex changes. This implies that the expression of the inciting angiogenic factors are hormone dependent. Recently, a second family of vascular endothelial growth factors was identified, the angiopoietins. Angiopoietins are vascular endothelial cell-specific growth factors that play important roles principally during the later stages of angiogenesis, after the induction of new capillaries by vascular endothelial growth factor (VEGF). There are four known angiopoietins, and their specificity for the vascular endothelium results from the restricted expression pattern of their tyrosine kinase receptor, Tie2. In this review, we discuss the molecular characterization and mechanism of action of angiopoietin-1 and angiopoietin-2 in reproductive tract angiogenesis. TARGET AUDIENCE: Obstetricians & Gynecologists, Family Physicians LEARNING OBJECTIVES: After completion of this article, the reader will be able to describe the angiogenic process and specifically explain the role of angiopoietics in reproductive tract angiogenesis and compare the differences between the various proteins that are involved in angiogenesis.


Subject(s)
Embryonic Structures/blood supply , Genitalia, Female/blood supply , Membrane Glycoproteins/physiology , Neovascularization, Physiologic/physiology , Proteins/physiology , Proto-Oncogene Proteins , Angiopoietin-1 , Angiopoietin-2 , Animals , Female , Humans , Neoplasm Proteins , Neovascularization, Pathologic/physiopathology , Pregnancy , Receptor, TIE-2
15.
Obstet Gynecol Surv ; 55(7 Suppl 2): S21-47, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10890575

ABSTRACT

Estrogen replacement therapy has significant potential benefits for postmenopausal women, such as improvement of menopausal symptoms and protection from osteoporosis, but it may also increase a woman's risk of breast cancer. Also, some women do not take hormone replacement therapy because of such undesirable side effects as breast tenderness and uterine bleeding. Therefore, there is much interest in the development of compounds that provide the benefits of estrogen replacement therapy without the risks and side effects. The selective estrogen receptor modulators make up one class of compounds with both estrogen agonist and antagonist activity. This review discusses the clinical indications, risks, benefits, and mechanisms of action of selective estrogen receptor modulators and related compounds.


Subject(s)
Estradiol/analogs & derivatives , Estrogen Replacement Therapy , Postmenopause , Selective Estrogen Receptor Modulators , Fulvestrant , Genistein , Humans , Raloxifene Hydrochloride , Tamoxifen
16.
Ann N Y Acad Sci ; 905: 177-87, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10818453

ABSTRACT

The specificity of cellular effects of lysolipid phosphate (LLP) growth factors is determined by binding to endothelial differentiation gene-encoded G protein-coupled receptors (EDG Rs), which transduce diverse proliferative and effector signals. The primary determinants of cellular responses to LLPs are the generative and biodegradative events, which establish steady-state concentrations of each LLP at cell surfaces, and the relative frequency of expression of each EDG R. There are major differences among types of cells in the net effective generation of the LLPs, lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), and in their profile of expression of EDG Rs. The less well characterized secondary determinants of cellular specificity of LLPs are high-affinity binding proteins with carrier and cell-presentation functions, cell-selective regulators of expression of EDG Rs, and cellular factors that govern coupling of EDG Rs to G protein transductional pathways. The roles of components of the LLP-EDG R system in normal physiology and disease processes will be definitively elucidated only after development of animal models with biologically meaningful alterations in genes encoding EDG Rs and the discovery of potent and selective pharmacological probes.


Subject(s)
Cell Division/physiology , Cell Survival/physiology , Lysophospholipids/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , GTP-Binding Proteins/metabolism , Humans , Lysophospholipids/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Cell Surface/metabolism , Tumor Cells, Cultured
17.
Ann N Y Acad Sci ; 905: 188-208, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10818454

ABSTRACT

Levels of lysophosphatidic acid (LPA) and lysophosphatidylcholine (LPC) are elevated in the plasma and ascites of ovarian cancer patients, but not in most other tumor types. LPA increases cell proliferation, cell survival, resistance to cisplatin, cell shrinkage, and production of vascular endothelial growth factor, urokinase plasminogen activator, and LPA itself in ovarian cancer cells, but not in normal ovarian surface epithelial cells. PSP24 and members of the endothelial differentiation gene (EDG) family (EDG1, EDG2, EDG4, and EDG7) of G protein-coupled receptors mediate LPA signaling. Ovarian cancer cell lines do not express EDG1 mRNA, have variable EDG2 mRNA and protein levels, and frequently exhibit levels of EDG4 mRNA and protein, suggesting that EDG4 may contribute to the deleterious effects of LPA in ovarian cancer. In contrast, activation of the EDG2 LPA receptor on ovarian cancer cells may lead to apoptosis and counter the effects of other LPA receptors. Thus, the development of agonists and antagonists for the appropriate spectrum of LPA receptors may alter proliferation, apoptosis, or response to therapy of ovarian cancer cells. Indeed, over 60% of all current drugs target the G protein-coupled family of receptors, making the LPA receptor family a "drugable" target. LPC, although not as thoroughly studied, increases cellular proliferation and mediates multiple other functions through unique signaling pathways.


Subject(s)
Growth Substances/physiology , Lysophospholipids/physiology , Ovarian Neoplasms/pathology , Ascites/metabolism , Female , Gene Expression Regulation/physiology , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/metabolism , Neoplasm Metastasis , Ovarian Neoplasms/therapy , Ovary/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction
18.
Semin Perinatol ; 24(1): 79-81, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10709866

ABSTRACT

The cyclic angiogenesis that occurs uniquely within the female reproductive tract is critical for normal reproduction. Two families of endothelial cell-specific growth factors and their receptors have been identified in the ovary, uterus, and placenta: vascular endothelial growth factor/vascular permeability factor and the angiopoietins. These appear to have complementary actions on the vasculature and to be involved during intrauterine development as well as in the adult. Within the ovary, a complex cascade of events required for angiogenesis may play a role in follicular maturation and selection as well as in normal corpus luteum function. Aberrant expression of angiogenic factors plays a role in a wide variety of abnormalities in the ovary. In the uterus, angiogenesis is required for endometrial growth and remodeling. Peptide growth factors such as vascular endothelial growth factor may serve as local mediators of the effects of reproductive hormones on the endometrial vasculature. Disease states such as dysfunctional uterine bleeding, endometriosis, and endometrial hyperplasia or cancer may be associated with aberrant uterine angiogenesis.


Subject(s)
Neovascularization, Physiologic/physiology , Reproduction/physiology , Animals , Female , Humans
19.
Clin Cancer Res ; 6(3): 880-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741711

ABSTRACT

Phosphatidylinositol 3-kinase (PI3-K) induces mitogenesis, cell growth, and cell transformation. Amplification of the gene encoding the P110alpha subunit likely is an important event in ovarian cancer progression, and PI3-K inhibitors are possible therapeutic agents for this disease. We evaluated effects of LY294002, a potent inhibitor of PI3-K, on growth of ovarian carcinoma in vivo and in vitro, and on ascites formation in vivo. Athymic mice were inoculated i.p. with the ovarian cancer cell line OVCAR-3. Seven days after inoculation, mice were treated with or without LY294002 (100 mg/kg of body weight) for 3 weeks. Body weight and abdominal circumference were measured twice weekly. At the end of the experiment, mice were sacrificed, ascites volume was measured, and tumors were excised. Mean tumor burden in the LY294002-treated group was reduced by approximately 65% versus controls. Virtually no ascites developed in the treatment group; mean volume of ascites in controls was 3.3 +/- 0.38 ml. OVCAR-3 cells also were cultured in vitro without and with LY294002 (1, 5, and 10 microM) for 24 h. The number of cells in 1, 5, and 10 microM LY294002-treated wells was reduced by 27, 56, and 75%, respectively, versus controls. In vivo and in vitro morphological studies demonstrated that LY294002 induced marked nuclear pyknosis and diminished cytoplasmic volume in the tumor cells, confirmed as apoptosis. Thus, LY294002 significantly inhibits growth and ascites formation of ovarian carcinoma in vivo and markedly inhibits ovarian cancer cell proliferation in vitro, suggesting an important role of PI3-K inhibitors as a potentially useful treatment for women with ovarian carcinoma.


Subject(s)
Cell Division/drug effects , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Ovarian Neoplasms/prevention & control , Phosphoinositide-3 Kinase Inhibitors , Animals , Apoptosis/drug effects , Ascites/pathology , Ascites/prevention & control , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
20.
J Clin Endocrinol Metab ; 84(10): 3732-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523022

ABSTRACT

CRH directly stimulates dehydroepiandrosterone sulfate (DHEAS) production in human fetal adrenal cells. In the human fetal and adult pituitary, CRH acts via protein kinase A (PKA). We determined the CRH signal transduction pathway in fetal adrenal cells, i.e. whether CRH modulates human fetal adrenal steroidogenesis via PKA and/or protein kinase C (PKC). In primary cultures, CRH increased inositol trisphosphate. After CRH treatment, inositol tris-, bis-, and monophosphates increased within 1 min, reaching maximal levels at 5 min. In contrast, PGF2alpha, known to act via PKC, induced a sustained response for up to 20 min. The response to CRH was dose dependent, maximal at 1 micromol/L at both 1 and 5 min. CRH increased DHEAS production, with a much lesser effect on cortisol. CRH did not stimulate inositol phospholipid in adult adrenal glands, suggesting that this pathway is unique to the fetal adrenal. CRH increased messenger ribonucleic acid encoding 17alpha-hydroxylase/17,20 lyase (P450c17), but not 3beta-hydroxysteroid dehydrogenase/delta(4-5) isomerase. However, 3betaHSD expression was stimulated by ACTH. PKC, but not PKA, inhibitors blocked CRH-stimulated P450c17 induction, whereas PKA inhibitors blocked ACTH-stimulated cortisol. Thus, CRH is coupled to the phospholipase C-inositol phosphate second messenger system and preferentially induces the expression of P450c17 and DHEAS, suggesting a unique role of CRH regulating human fetal adrenal function via PKC.


Subject(s)
Adrenal Glands/embryology , Corticotropin-Releasing Hormone/pharmacology , Fetus/metabolism , Protein Kinase C/physiology , Steroid 17-alpha-Hydroxylase/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Adrenocorticotropic Hormone/pharmacology , Cyclic AMP/biosynthesis , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dinoprost/pharmacology , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Fetus/cytology , Humans , Inositol Phosphates/metabolism , Protein Kinase C/antagonists & inhibitors , Steroids/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...