Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
ACS Biomater Sci Eng ; 7(7): 3209-3220, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34160196

ABSTRACT

Successful treatment of glioblastoma (GBM) is hampered by primary tumor recurrence after surgical resection and poor prognosis, despite adjuvant radiotherapy and chemotherapy. In search of improved outcomes for this disease, quisinostat appeared as a lead compound in drug screening. A delivery system was devised for this drug and to exploit current clinical methodology: an injectable hydrogel, loaded with both the quisinostat drug and radiopaque gold nanoparticles (AuNP) as contrast agent, that can release these payloads as a response to radiation. This hydrogel grants high local drug concentrations, overcoming issues with current standards of care. Significant hydrogel degradation and quisinostat release were observed due to the radiation trigger, providing high in vitro anticancer activity. In vivo, the combination of radiotherapy and the radiation-induced delivery of quisinostat from the hydrogel, successfully inhibited tumor growth in a mice model bearing xenografted human GBM tumors with a total response rate of 67%. Long-term tolerability was observed after intratumoral injection of the quisinostat loaded hydrogel. The AuNP payload enabled precise image-guided radiation delivery and the monitoring of hydrogel degradation using computed tomography (CT). These exciting results highlight this hydrogel as a versatile imageable drug delivery platform that can be activated simultaneously to radiation therapy and potentially offers improved treatment for GBM.


Subject(s)
Glioblastoma , Metal Nanoparticles , Glioblastoma/diagnostic imaging , Gold , Humans , Hydrogels , Neoplasm Recurrence, Local
2.
Cell ; 180(1): 188-204.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31883794

ABSTRACT

Glioblastomas exhibit vast inter- and intra-tumoral heterogeneity, complicating the development of effective therapeutic strategies. Current in vitro models are limited in preserving the cellular and mutational diversity of parental tumors and require a prolonged generation time. Here, we report methods for generating and biobanking patient-derived glioblastoma organoids (GBOs) that recapitulate the histological features, cellular diversity, gene expression, and mutational profiles of their corresponding parental tumors. GBOs can be generated quickly with high reliability and exhibit rapid, aggressive infiltration when transplanted into adult rodent brains. We further demonstrate the utility of GBOs to test personalized therapies by correlating GBO mutational profiles with responses to specific drugs and by modeling chimeric antigen receptor T cell immunotherapy. Our studies show that GBOs maintain many key features of glioblastomas and can be rapidly deployed to investigate patient-specific treatment strategies. Additionally, our live biobank establishes a rich resource for basic and translational glioblastoma research.


Subject(s)
Cell Culture Techniques/methods , Glioblastoma/metabolism , Organoids/growth & development , Adult , Aged , Aged, 80 and over , Animals , Biological Specimen Banks , Female , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Models, Biological , Organoids/metabolism , Reproducibility of Results , Xenograft Model Antitumor Assays/methods
3.
Neurooncol Adv ; 1(1): vdz030, 2019.
Article in English | MEDLINE | ID: mdl-31807732

ABSTRACT

BACKGROUND: Amplification of the epidermal growth factor receptor (EGFR) gene is commonly found in glioblastoma (GBM). About 57% GBM overexpresses EGFR and are associated with tumor progression, poor prognosis, and shorter life expectancy. Molecular profiling of solid tumors usually takes several weeks and may be biased by intrinsic tumor heterogeneity. METHODS: The unique sequence created by the fusion of exon 1 and exon 8 in EGFRvIII was used to guide the design of primers and a Minor Groove Binder (MGB) probe. Extracted total RNA was reverse transcribed and pre-amplified by PCR, followed by detection of the EGFRvIII mutation by dPCR. RESULTS: The lowest limit of quantification of our EGFRvIII assay was 0.003%. The EGFRvIII variant was identified in patient-derived glioma neurosphere cell lines, xenograft mouse model, and patient-derived tumor specimens. The overall workflow can be accomplished within 24 hours. In certain samples, EGFRvIII was detected when next-generation sequencing was unable to identify the variant. This finding highlights the ability of the dPCR assay to identify EGFRvIII mutations in heterogeneous solid tumors such as GBM in a rapid fashion by profiling samples from spatially distinct areas of tumors from the same patient. CONCLUSIONS: In this study, we developed a highly sensitive digital PCR (dPCR) platform and leveraged our assay to detect the variant III alteration of EGFR (EGFRvIII) and amplified EGFR in patient-derived glioma neurosphere cell lines, orthotopic xenograft GBM mouse models, and patient-derived tumor specimens in less than 24 hours from minute quantities of starting material.

4.
CNS Oncol ; 8(3): CNS43, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31769726

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and carries a dismal prognosis. The EGFR gene is among the most commonly deranged genes in GBM and thus an important therapeutic target. We report the case of a young female with heavily pretreated EGFR-mutated GBM, for whom we initiated osimertinib, an oral, third-generation tyrosine kinase inhibitor that irreversibly inhibits EGFR and has significant brain penetration. We then review some of the main challenges in targeting EGFR, including lack of central nervous system penetration with most tyrosine kinase inhibitors, molecular heterogeneity of GBM and the need for enhanced specificity for the EGFR mutations relevant in GBM.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Mutation , Protein Kinase Inhibitors/therapeutic use , Adult , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Prognosis
5.
Curr Pharm Biotechnol ; 20(14): 1163-1171, 2019.
Article in English | MEDLINE | ID: mdl-31433750

ABSTRACT

Blood Brain Barrier (BBB) is the collection of vessels of blood with special properties of permeability that allow a limited range of drug and compounds to pass through it. The BBB plays a vital role in maintaining balance between intracellular and extracellular environment for brain. Brain Capillary Endothelial Cells (BECs) act as vehicle for transport and the transport mechanisms across BBB involve active and passive diffusion of compounds. Efficient prediction models of BBB permeability can be vital at the preliminary stages of drug development. There have been persistent efforts in identifying the prediction of BBB permeability of compounds employing multiple machine learning methods in an attempt to minimize the attrition rate of drug candidates taking up preclinical and clinical trials. However, there is an urgent need to review the progress of such machine learning derived prediction models in the prediction of BBB permeability. In the current article, we have analyzed the recently developed prediction model for BBB permeability using machine learning.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability/physiology , Endothelial Cells/metabolism , Machine Learning , Models, Biological , Animals , Brain/metabolism , Diffusion , Drug Development , Humans , Permeability , Pharmaceutical Preparations/metabolism
6.
Mol Cancer Res ; 17(5): 1102-1114, 2019 05.
Article in English | MEDLINE | ID: mdl-30642878

ABSTRACT

The high incidence of glioblastoma recurrence necessitates additional therapeutic strategies. Heterogeneous populations of cells, including glioma stem cells (GSC) have been implicated in disease recurrence. GSCs are able to survive irradiation and temozolomide (TMZ) treatment due to upregulation of DNA damage pathways. One potential strategy to target treatment-resistant tumor populations may be via the integrated stress response (ISR). Modulation of the ISR pathway also allows for sensitization of treatment-resistant cells to TRAIL. We generated a novel cell-based death receptor assay to identify potent inducers of ISR-dependent DR5 expression. We used this assay to screen compounds from three commercially available libraries, and identified 1-benzyl-3-cetyl-2-methylimidazolium iodide (NH125) as a potent inducer of DR5 expression. NH125 engages the EIF2α-ATF4-CHOP axis culminating in DR5 expression at low micromolar doses. Expression of CHOP plays a critical role in NH125-mediated TRAIL synergy. Treatment of GSC with NH125 produces a marked reduction in viability when compared with other cell lines. NH125-treated GSC also synergize with lower doses of TRAIL when compared with all other cell lines tested. Transcriptional analysis of NH125-treated GSC uncovers a unique profile that involves activation of ISR and GADD45 pathways. Treatment of GSC xenografts with encapsulated PEG-PCL-NH125 leads to a sustained decrease in tumor volume. IMPLICATIONS: Taken together, these data suggest that engaging the ISR pathway represents a promising strategy to target treatment refractory GSC that have been implicated in glioblastoma recurrence.


Subject(s)
Brain Neoplasms/genetics , Glioma/drug therapy , Imidazoles/administration & dosage , Neoplastic Stem Cells/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/metabolism , Humans , Imidazoles/pharmacology , Mice , Neoplastic Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transcription Factor CHOP/genetics , Xenograft Model Antitumor Assays
7.
Cancer Res ; 78(23): 6632-6642, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30322863

ABSTRACT

: Circulating tumor cells (CTC) are known to be present in the blood of patients with glioblastoma (GBM). Here we report that GBM-derived CTC possess a cancer stem cell (CSC)-like phenotype and contribute to local tumorigenesis and recurrence by the process of self-seeding. Genetic probes showed that mouse GBM-derived CTC exhibited Sox2/ETn transcriptional activation and expressed glioma CSC markers, consistent with robust expression of stemness-associated genes including SOX2, OCT4, and NANOG in human GBM patient-derived samples containing CTC. A transgenic mouse model demonstrated that CTC returned to the primary tumor and generated new tumors with enhanced tumorigenic capacity. These CTCs were resistant to radiotherapy and chemotherapy and to circulation stress-induced cell apoptosis. Single-cell RNA-seq analysis revealed that Wnt activation induced stemness and chemoresistance in CTC. Collectively, these findings identify GBM-derived CTC as CSC-like cells and suggest that targeting Wnt may offer therapeutic opportunities for eliminating these treatment-refractory cells in GBM. SIGNIFICANCE: These findings identify CTCs as an alternative source for in situ tumor invasion and recurrence through local micrometastasis, warranting eradication of systemic "out-of-tumor" CTCs as a promising new therapeutic opportunity for GBM.


Subject(s)
Glioma/metabolism , Glioma/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm , Female , Humans , Immunophenotyping , Male , Mice , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phenotype , Stress, Physiological , Wnt Proteins/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L168-74, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25934671

ABSTRACT

Developmental mechanisms leading to lung hypoplasia in congenital diaphragmatic hernia (CDH) remain poorly defined. Pulmonary innervation is defective in the human disease and in the rodent models of CDH. We hypothesize that defective parasympathetic innervation may contribute to airway branching abnormalities and, therefore, lung hypoplasia, during lung development in CDH. The murine nitrofen model of CDH was utilized to study the effect of the cholinergic agonist carbachol on embryonic day 11.5 (E11.5) lung explant cultures. Airway branching and contractions were quantified. In a subset of experiments, verapamil was added to inhibit airway contractions. Sox9 immunostaining and 5-bromo-2-deoxyuridine incorporation were used to identify and quantify the number and proliferation of distal airway epithelial progenitor cells. Intra-amniotic injections were used to determine the in vivo effect of carbachol. Airway branching and airway contractions were significantly decreased in nitrofen-treated lungs compared with controls. Carbachol resulted in increased airway contractions and branching in nitrofen-treated lungs. Nitrofen-treated lungs exhibited an increased number of proliferating Sox9-positive distal epithelial progenitor cells, which were decreased and normalized by treatment with carbachol. Verapamil inhibited the carbachol-induced airway contractions in nitrofen-treated lungs but had no effect on the carbachol-induced increase in airway branching, suggesting a direct carbachol effect independent of airway contractions. In vivo treatment of nitrofen-treated embryos via amniotic injection of carbachol at E10.5 resulted in modest increases in lung size and branching at E17.5. These results suggest that defective parasympathetic innervation may contribute to airway branching abnormalities in CDH.


Subject(s)
Embryo, Mammalian/pathology , Hernias, Diaphragmatic, Congenital/pathology , Lung/abnormalities , Lung/pathology , Parasympathetic Nervous System/pathology , Respiratory System/pathology , Animals , Calcium Channel Blockers/pharmacology , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , Disease Models, Animal , Embryo, Mammalian/drug effects , Female , Hernias, Diaphragmatic, Congenital/chemically induced , Hernias, Diaphragmatic, Congenital/embryology , Humans , Immunoenzyme Techniques , Lung/drug effects , Mice , Parasympathetic Nervous System/embryology , Parasympathetic Nervous System/metabolism , Pesticides/toxicity , Phenyl Ethers/toxicity , Respiratory System/drug effects , Respiratory System/embryology , Stem Cells/drug effects , Verapamil/pharmacology
9.
PLoS One ; 10(3): e0123376, 2015.
Article in English | MEDLINE | ID: mdl-25807549

ABSTRACT

BACKGROUND: Circulating tumor cell (CTC) detection and genetic analysis may complement currently available disease assessments in patients with melanoma to improve risk stratification and monitoring. We therefore sought to establish the feasibility of a telomerase-based assay for detecting and isolating live melanoma CTCs. METHODS: The telomerase-based CTC assay utilizes an adenoviral vector that, in the presence of elevated human telomerase activity, drives the amplification of green fluorescent protein. Tumor cells are then identified via an image processing system. The protocol was tested on melanoma cells in culture or spiked into control blood, and on samples from patients with metastatic melanoma. Genetic analysis of the isolated melanoma CTCs was then performed for BRAF mutation status. RESULTS: The adenoviral vector was effective for all melanoma cell lines tested with sensitivity of 88.7% (95%CI 85.6-90.4%) and specificity of 99.9% (95%CI 99.8-99.9%). In a pilot trial of patients with metastatic disease, CTCs were identified in 9 of 10 patients, with a mean of 6.0 CTCs/mL. At a cutoff of 1.1 CTCs/mL, the telomerase-based assay exhibits test performance of 90.0% sensitivity and 91.7% specificity. BRAF mutation analysis of melanoma cells isolated from culture or spiked control blood, or from pilot patient samples was found to match the known BRAF mutation status of the cell lines and primary tumors. CONCLUSIONS: To our knowledge, this is the first report of a telomerase-based assay effective for detecting and isolating live melanoma CTCs. These promising findings support further studies, including towards integrating into the management of patients with melanoma receiving multimodality therapy.


Subject(s)
Melanoma/pathology , Neoplastic Cells, Circulating/metabolism , Adenoviridae/genetics , Adult , Aged , Area Under Curve , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Linear Models , Male , Melanoma/metabolism , Middle Aged , Mutation , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Pilot Projects , Proto-Oncogene Proteins B-raf/genetics , ROC Curve , Telomerase/metabolism
10.
Endocrinology ; 148(2): 726-34, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17095585

ABSTRACT

Granulosa cells express the closely related orphan nuclear receptors steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1). To determine whether SF-1 and LRH-1 have differential effects on steroid production, we compared the effects of overexpressing LRH-1 and SF-1 on estrogen and progesterone production by undifferentiated rat granulosa cells. Adenovirus mediated overexpression of LRH-1 or SF-1 had qualitatively similar effects. Neither LRH-1 nor SF-1 alone stimulated estrogen or progesterone production, but when combined with FSH and testosterone, each significantly augmented progesterone production and mRNAs for cholesterol side-chain cleavage enzyme and 3beta-hydroxysteroid dehydrogenase above that observed with FSH alone, with SF-1 being more effective than LRH-1. LRH-1 did not augment FSH-stimulated estrogen production, whereas SF-1 produced only a slight ( approximately 30%) augmentation of FSH-stimulated estrogen production. The stimulatory actions of both were reduced by overexpression of dosage-sensitive sex reversal, adrenal hypoplasia congenita, critical region on the X chromosome, gene 1. Expression of either LRH-1 or SF-1 together with constitutively active protein kinase B in the absence of FSH stimulated progesterone production and mRNAs for 3beta-hydroxysteroid dehydrogenase and cholesterol side-chain cleavage enzyme but did not stimulate estrogen production or mRNA for aromatase. These findings demonstrate that LRH-1 and SF-1 have qualitatively similar actions on FSH-stimulated estrogen and progesterone production, which would suggest that these factors may have overlapping actions in the regulation of steroidogenesis that accompanies granulosa cell differentiation.


Subject(s)
Granulosa Cells/metabolism , Homeodomain Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Steroids/biosynthesis , Transcription Factors/physiology , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Drug Interactions , Drug Synergism , Estrogens/biosynthesis , Female , Follicle Stimulating Hormone/pharmacology , Gene Transfer Techniques , Granulosa Cells/drug effects , Homeodomain Proteins/genetics , Humans , Mice , Progesterone/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , RNA, Messenger/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Steroidogenic Factor 1 , Time Factors , Transcription Factors/genetics
11.
FEBS Lett ; 580(24): 5653-60, 2006 Oct 16.
Article in English | MEDLINE | ID: mdl-17011554

ABSTRACT

Establishment of early pregnancy is promoted by a complex network of signalling molecules that mediate cell-to-cell and cell-to-extracellular matrix communications between the receptive endometrium and the invasive trophectoderm. In this study, we have attempted to evaluate the expression profiles of cadherin and catenin during embryo implantation in the mouse. Western blotting studies along with immunocytochemical analysis revealed that E-cadherin is expressed rather ubiquitously in the uterine epithelial cells, distinct enrichment is observed on the apical membrane in the endometrium of peri-implantation uterus specifically at the implantation sites and not at the inter-implanation sites. beta-Catenin also is upregulated and is specifically restricted to apical membrane of epithelial cells of implantation sites. Progesterone induced expression of E-cadherin and 17beta-estradiol regulated the expression of catenin in implantation-delayed uteri. Interestingly, estradiol imparted negative modulation on cadherin expression when co-administered with progesterone. On the contrary, trophoblast exhibits a striking down regulation of cadherin, catenin and Ca(2+) at peri implanting stage. These observations suggest that the trophoblasts exhibited an invasive phenotype while the endometrial epithelium displayed an adhesive phenotype during the window of implantation. Thus, embryo implantation presents an instance where two interacting surfaces showed mutually complementing interaction phenotypes.


Subject(s)
Cadherins/metabolism , Calcium/metabolism , Embryo Implantation , Embryo, Mammalian/physiology , Uterus/physiology , beta Catenin/metabolism , Animals , Embryo, Mammalian/metabolism , Female , Mice , Time Factors , Uterus/metabolism
12.
Endocrinology ; 145(8): 3821-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15117876

ABSTRACT

FSH-stimulated granulosa cell differentiation is associated with the induction of the LH receptor (LHr) as well as induction of the estrogen and progesterone biosynthetic pathways. Although activation of the cAMP-protein kinase A pathway is sufficient to stimulate progesterone production, additional pathways are required for the induction of the LHr and p450 aromatase. The orphan nuclear receptor, liver receptor homolog-1 (LRH-1), is expressed in granulosa cells and has been shown to synergize with the cAMP signaling system to regulate the gonadal type II aromatase promoter in transient transfection assays. To determine whether LRH-1 can interact with the cAMP pathway in the induction of aromatase and the LHr, we examined the effects of an adenoviral vector that directs the expression of human LRH-1 (Ad-LRH-1) on FSH-stimulated granulosa cell differentiation. Infection of undifferentiated granulosa cells with LRH-1 alone had no effect on estrogen production, progesterone production, or the expression of the LHr. However, combination of FSH stimulation and Ad-LRH-1 infection led to significantly greater progesterone production and increases in mRNA for p450 side-chain cleavage and 3beta-hydroxysteroid dehydrogenase than granulosa cells stimulated by FSH alone. However, infection with Ad-LRH-1 did not stimulate estradiol production or increases in mRNA for p450 aromatase or the LHr above that seen with FSH treatment alone. Moreover, infection with Ad-LRH-1 was able to overcome H-89 inhibition of FSH-stimulated progesterone but not estrogen production. Collectively, these observations support a direct role for LRH-1 in the induction of the progesterone but not the estrogen biosynthetic pathway during granulosa cell differentiation.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Progesterone/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenoviridae/genetics , Animals , Cell Differentiation , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/physiology , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/physiology , Dose-Response Relationship, Drug , Estrogens/biosynthesis , Female , Granulosa Cells/metabolism , Humans , Rats , Receptors, Retinoic Acid/physiology , Repressor Proteins/physiology
13.
Endocrinology ; 144(9): 3985-94, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12933673

ABSTRACT

Although FSH receptors are linked to the cAMP second messenger system, additional intracellular signaling pathways appear to be required for the induction of aromatase and the LH receptor during granulosa cell differentiation. We employed adenovirus vectors to modulate specific intracellular signaling systems in undifferentiated granulosa cells to identify the signaling pathway(s) that may be involved in the FSH-mediated induction of aromatase and the LH receptor. Expression of either the constitutively activated human LH receptor D578H or the constitutively active human G(s)alpha Q227L resulted in increased cAMP production without increasing aromatase activity or mRNA levels for the LH receptor. To explore the contributions of other pathways, we expressed the constitutively activated forms MAPK kinase (MEK) and protein kinase B (PKB). Neither MEK nor PKB alone increased estrogen or progesterone production by undifferentiated granulosa cells. Stimulation of granulosa cells by FSH in the presence of the constitutively active PKB, but not MEK, led to an amplification of FSH-induced aromatase and LH receptor mRNA levels, whereas a dominant negative PKB vector completely abolished the actions of FSH. The expression of the constitutively active PKB in combination with the constitutively active LH receptor D578H, the constitutively active G(s)alpha Q227L, or 8-bromo-cAMP led to an induction of aromatase as well as LH receptor mRNA comparable to that seen in cells stimulated with FSH alone. These results demonstrate that PKB is an essential component of the FSH-mediated granulosa cell differentiation and that both PKB and G(s)alpha signaling pathways are required.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Granulosa Cells/cytology , Granulosa Cells/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sulfonamides , Adenoviridae/genetics , Androstadienes/pharmacology , Aromatase/metabolism , Biomarkers , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/biosynthesis , Cyclic AMP/pharmacology , Enzyme Inhibitors/pharmacology , Estrogens/biosynthesis , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Granulosa Cells/drug effects , Humans , Isoquinolines/pharmacology , Progesterone/biosynthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...