Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 8(1): 134-40, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22099027

ABSTRACT

Disease-specific induced pluripotent stem cells (iPSC) allow unprecedented experimental platforms for basic research as well as high-throughput screening. This may be particularly relevant for neuropsychiatric disorders, in which the affected neuronal cells are not accessible. Keratinocytes isolated from hair follicles are an ideal source of patients' cells for reprogramming, due to their non-invasive accessibility and their common neuroectodermal origin with neurons, which can be important for potential epigenetic memory. From a small number of plucked human hair follicles obtained from two healthy donors we reprogrammed keratinocytes to pluripotent iPSC. We further differentiated these hair follicle-derived iPSC to neural progenitors, forebrain neurons and functional dopaminergic neurons. This study shows that human hair follicle-derived iPSC can be differentiated into various neural lineages, suggesting this experimental system as a promising in vitro model to study normal and pathological neural developments, avoiding the invasiveness of commonly used skin biopsies.


Subject(s)
Cell Differentiation , Hair Follicle/cytology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Nervous System Diseases/pathology , Animals , Cell Differentiation/genetics , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neurons/cytology , Neurons/metabolism
2.
Cell Death Differ ; 16(5): 728-37, 2009 May.
Article in English | MEDLINE | ID: mdl-19219066

ABSTRACT

After genotoxic stress, normal cells trigger DNA repair or, if unable to repair, undergo apoptosis to eradicate the cells that bear the risk of becoming tumorigenic. Here we show that repression of the transcription factor, activating transcription factor 3 (ATF3), after ultraviolet (UV)-mediated genotoxic stress impairs the DNA repair process. We provide evidence that ATF3 directly regulates the proliferating cell nuclear antigen (PCNA)-associated factor KIAA0101/p15(PAF). We further show that the expressions of ATF3 and p15(PAF) is sufficient to trigger the DNA repair machinery, and that attenuation of their expression alters DNA repair mechanisms. We show that the expression of p15(PAF) compensates for a lack of ATF3 expression, thereby constituting a major effector of ATF3 in the DNA repair process. In addition, we provide evidence that p15(PAF) expression is required for the correct function of PCNA during DNA repair, as prevention of their interaction significantly alters DNA repair mechanisms. Finally, defective DNA repair, because of the downregulation of p15(PAF) expression, rendered the cells more sensitive to UV-induced cell death. Therefore, our results suggest ATF3 and p15(PAF) as novel gatekeepers of genomic integrity after UV exposure.


Subject(s)
Activating Transcription Factor 3/metabolism , Carrier Proteins/metabolism , DNA Repair , Apoptosis , Cell Line , DNA Damage , DNA-Binding Proteins , Down-Regulation , Genome , HeLa Cells , Humans , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Ultraviolet Rays
3.
Cell Death Differ ; 15(9): 1472-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511933

ABSTRACT

In this study, we describe a novel activating transcription factor 3 (ATF3)-dependent death pathway triggered by ultraviolet (UV) irradiation. We demonstrate that ATF3 contributes to UV-induced apoptosis through the regulation of hypoxia inducible factor (Hif)-2alpha expression, which in turn induces the expression of proapoptotic genes, such as Caspase7 or TRAIL (tumor necrosis factor (ligand) superfamily, member 10). Gain of function of Hif-2alpha as well as ATF3 is sufficient to trigger cell death, whereas loss of function of both proteins drastically inhibits UV-induced apoptosis. Repression of Hif-2alpha strongly impairs ATF3-mediated death, providing evidences that Hif-2alpha is the major death effector of ATF3. In addition, Hif-1alpha, already known as a proapoptotic gene, upon UV irradiation, is not able to compensate for the lack of Hif-2alpha expression, thereby confirming the major contribution of Hif-2alpha in UV-mediated cell death. We further demonstrate that this cascade of gene activation depends on p38 and c-Jun N-terminal kinase (JNK) activity. Impairment of such a pathway is likely to contribute to oncogenesis by promoting survival of cells that could accumulate severe chromosomal alterations.


Subject(s)
Activating Transcription Factor 3/physiology , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ultraviolet Rays , Activating Transcription Factor 3/biosynthesis , Activating Transcription Factor 3/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , RNA, Messenger/metabolism , Signal Transduction
4.
Cell Death Differ ; 13(7): 1075-87, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16311513

ABSTRACT

Embryonic ectoderm is fated to become either neural or epidermal, depending on patterning processes that occur before and during gastrulation. It has been stated that epidermal commitment proceeds from a bone morphogenetic protein-4 (BMP-4)-dependent inhibition of dorsal ectoderm neuralization. We recently demonstrated that murine embryonic stem (ES) cells treated with BMP-4 undergo effective keratinocyte commitment and epidermogenesis. Focusing on the precise role of BMP-4 in the early choice between neural and epidermal commitment, we show here that BMP-4 treatment of ES cells leads to a dramatic apoptotic death of Sox-1+ neural precursors with concomitant epidermal engagement. In addition, neutralization of the Smad pathway prevents both the BMP-4 apoptotic process and the inhibition of neural differentiation. Our results suggest that, in mammals, BMP-4, as an active inducer of epidermal commitment, interferes with the survival of neural precursors through induction of their apoptotic cell death.


Subject(s)
Apoptosis/drug effects , Bone Morphogenetic Proteins/pharmacology , Neurons/drug effects , Smad6 Protein/physiology , Stem Cells/drug effects , 3T3 Cells , Animals , Apoptosis/physiology , Bone Morphogenetic Protein 4 , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fluorescent Antibody Technique , Gene Expression/drug effects , Mice , Microscopy, Confocal , Neurons/cytology , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Smad6 Protein/genetics , Smad6 Protein/metabolism , Smad6 Protein/pharmacology , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Cell Biol ; 152(4): 843-50, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11266474

ABSTRACT

Normal pigmentation depends on the uniform distribution of melanin-containing vesicles, the melanosomes, in the epidermis. Griscelli syndrome (GS) is a rare autosomal recessive disease, characterized by an immune deficiency and a partial albinism that has been ascribed to an abnormal melanosome distribution. GS maps to 15q21 and was first associated with mutations in the myosin-V gene. However, it was demonstrated recently that GS can also be caused by a mutation in the Rab27a gene. These observations prompted us to investigate the role of Rab27a in melanosome transport. Using immunofluorescence and immunoelectron microscopy studies, we show that in normal melanocytes Rab27a colocalizes with melanosomes. In melanocytes isolated from a patient with GS, we show an abnormal melanosome distribution and a lack of Rab27a expression. Finally, reexpression of Rab27a in GS melanocytes restored melanosome transport to dendrite tips, leading to a phenotypic reversion of the diseased cells. These results identify Rab27a as a key component of vesicle transport machinery in melanocytes.


Subject(s)
Hypopigmentation/metabolism , Immunologic Deficiency Syndromes/metabolism , Melanocytes/metabolism , Melanosomes/metabolism , Myosin Type V , rab GTP-Binding Proteins/metabolism , Biological Transport , Calmodulin-Binding Proteins/isolation & purification , Cell Compartmentation , Fluorescent Antibody Technique , Humans , Melanocytes/ultrastructure , Melanosomes/ultrastructure , Microscopy, Immunoelectron , Molecular Motor Proteins , Nerve Tissue Proteins/isolation & purification , Syndrome , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/isolation & purification , rab GTP-Binding Proteins/ultrastructure , rab27 GTP-Binding Proteins
6.
J Invest Dermatol ; 115(3): 381-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10951272

ABSTRACT

Compelling evidence suggest a role for melanocortins in the regulation of melanogenesis by ultraviolet radiation. Within the epidermis, melanocytes and keratinocytes produce alpha-melanocyte-stimulating hormone and adrenocorticotropic hormone. The persistence and the strength of the biologic signal delivered by these peptides depend on their local concentration, which is controlled by the rate of peptide production and by the rate of its degradation. In this study, we investigated the mechanism of melanocortin degradation by melanocytes and the effect of ultraviolet on this process. We have focused our attention on a neutral endopeptidase, neprilysin, which has been implicated in the ending of numerous peptidergic signals. We have shown that this enzyme is expressed at the surface of human melanocytes. Interestingly, its activity and its expression are dramatically downregulated by ultraviolet B treatment. Moreover, in the presence of phosphoramidon, a stable inhibitor of neprilysin, we observed an increased efficiency of alpha-melanocyte-stimulating hormone and adrenocorticotropic hormone to stimulate both tyrosinase activity and microphthalmia expression. Taken together, these data indicate that neprilysin expressed by melanocytes has a physiologic role in the regulation of melanogenesis by proopiomelanocortin peptide. Further, its downregulation by ultraviolet B irradiation shed light on a new and appealing mechanism of ultraviolet B induced melanogenesis via the control of melanocortins degradation.


Subject(s)
Melanins/biosynthesis , Melanins/radiation effects , Neprilysin/radiation effects , Ultraviolet Rays , alpha-MSH/physiology , Cells, Cultured , Down-Regulation/radiation effects , Humans , Neprilysin/physiology , Skin Pigmentation/radiation effects
7.
EMBO J ; 19(12): 2900-10, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10856235

ABSTRACT

In melanocytes and melanoma cells, cAMP activates extracellular signal-regulated kinases (ERKs) and MEK-1 by an unknown mechanism. We demonstrate that B-Raf is activated by cAMP in melanocytes. A dominant-negative mutant of B-Raf, but not of Raf-1, blocked the cAMP-induced activation of ERK, indicating that B-Raf is the MEK-1 upstream regulator mediating this cAMP effect. Studies using Clostridium sordelii lethal toxin and Clostridium difficile toxin B have suggested that Rap-1 or Ras might transduce cAMP action. We show that Ras, but not Rap-1, is activated cell-specifically and mediates the cAMP-dependent activation of ERKs, while Rap-1 is not involved in this process in melanocytes. Our results suggest a novel, cell-specific mechanism involving Ras small GTPase and B-Raf kinase as mediators of ERK activation by cAMP. Also, in melanocytes, Ras or ERK activation by cAMP is not mediated through protein kinase A activation. Neither the Ras exchange factor, Son of sevenless (SOS), nor the cAMP-responsive Rap-1 exchange factor, Epac, participate in the cAMP-dependent activation of Ras. These findings suggest the existence of a melanocyte-specific Ras exchange factor directly regulated by cAMP.


Subject(s)
Cyclic AMP/metabolism , MAP Kinase Signaling System , Melanocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Kinase Kinases/metabolism , Melanoma, Experimental , Mice , Models, Biological , PC12 Cells , Proto-Oncogene Proteins c-raf/metabolism , Rats , Son of Sevenless Proteins/genetics , Son of Sevenless Proteins/metabolism , ras Proteins/genetics
8.
J Biol Chem ; 273(31): 19560-5, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9677380

ABSTRACT

In mouse follicular melanocytes, production of eumelanins (brown-black pigments) and pheomelanins (yellow-brownish pigments) is under the control of two intercellular signaling molecules that exert opposite actions, alpha-melanocyte-stimulating hormone (alphaMSH) which preferentially increases the synthesis of eumelanins, and agouti signal protein (ASP) whose expression favors the production of hair containing pheomelanins. In this study, we report that ASP does not only affect mature melanocytes but can also inhibit the differentiation of melanoblasts. We show that both alphaMSH and forskolin promote the differentiation of murine melanoblasts into mature melanocytes and that ASP inhibits this process. We present evidence that the expression of a specific melanogenic transcription factor, microphthalmia, and its binding to an M box regulatory element, is inhibited by ASP. We also show that, in B16 murine melanoma cells, ASP inhibits alphaMSH-stimulated expression of tyrosinase, tyrosine-related proteins 1 and 2 through an inhibition of the transcription activity of their respective promoters. Further, ASP inhibits alphaMSH-induced expression of the microphthalmia gene and reduces the level of microphthalmia in the cells. Our data demonstrate that ASP can regulate both melanoblast differentiation and melanogenesis, pointing out the key role of microphthalmia in the control of these processes.


Subject(s)
Cell Differentiation/drug effects , DNA-Binding Proteins/physiology , Intercellular Signaling Peptides and Proteins , Melanocytes/drug effects , Proteins/pharmacology , Transcription Factors , Agouti Signaling Protein , Animals , Colforsin/pharmacology , Cyclic AMP/pharmacology , Fungal Proteins/genetics , Gene Expression Regulation/drug effects , Melanins/biosynthesis , Melanoma/metabolism , Membrane Proteins/genetics , Mice , Microphthalmia-Associated Transcription Factor , Monophenol Monooxygenase/genetics , TRPC Cation Channels , Tumor Cells, Cultured , alpha-MSH/pharmacology
9.
Mol Cell Biol ; 18(2): 694-702, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9447965

ABSTRACT

In melanocytes and in melanoma cells, cyclic AMP (cAMP)-elevating agents stimulate melanogenesis and increase the transcription of tyrosinase, the rate-limiting enzyme in melanin synthesis. However, two other enzymes, tyrosinase-related protein 1 (TRP1) and TRP2, are required for a normal melanization process leading to eumelanin synthesis. In B16 melanoma cells, we demonstrated that stimulation of melanogenesis by cAMP-elevating agents results in an increase in tyrosinase, TRP1, and TRP2 expression. cAMP, through a cAMP-dependent protein kinase pathway, stimulates TRP1 and TRP2 promoter activities in both B16 mouse melanoma cells and normal human melanocytes. Regulation of the TRP1 and TRP2 promoters by cAMP involves a M box and an E box. Further, a classical cAMP response element-like motif participates in the cAMP responsiveness of the TRP2 promoter, demonstrating that the TRP2 gene is subjected to different regulatory processes, which could account for its different expression patterns during embryonic development or under specific physiological and pathological conditions. We also found that microphthalmia, a basic helix-loop-helix transcription factor, strongly stimulates the transcriptional activities of the TRP1 and TRP2 promoters, mainly through binding to the M boxes. Additionally, we demonstrated that cAMP increases microphthalmia expression and thereby its binding to TRP1 and TRP2 M boxes. These convergent and compelling results disclose at least a part of the molecular mechanism involved in the regulation of melanogenic gene expression by cAMP and emphasize the pivotal role of microphthalmia in this process.


Subject(s)
Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , Intramolecular Oxidoreductases/genetics , Leucine Zippers , Membrane Glycoproteins , Oxidoreductases , Promoter Regions, Genetic , Proteins/genetics , Transcription Factors/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Humans , Mice , Microphthalmia-Associated Transcription Factor , Monophenol Monooxygenase/metabolism , Tumor Cells, Cultured
10.
J Clin Invest ; 99(4): 635-42, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9045865

ABSTRACT

Ultraviolet (UV) radiation is the main physiological stimulus for human skin pigmentation. Within the epidermal-melanin unit, melanocytes synthesize and transfer melanin to the surrounding keratinocytes. Keratinocytes produce paracrine factors that affect melanocyte proliferation, dendricity, and melanin synthesis. In this report, we show that normal human keratinocytes secrete nitric oxide (NO) in response to UVA and UVB radiation, and we demonstrate that the constitutive isoform of keratinocyte NO synthase is involved in this process. Next, we investigate the melanogenic effect of NO produced by keratinocytes in response to UV radiation using melanocyte and keratinocyte cocultures. Conditioned media from UV-exposed keratinocytes stimulate tyrosinase activity of melanocytes. This effect is reversed by NO scavengers, suggesting an important role for NO in UV-induced melanogenesis. Moreover, melanocytes respond to NO-donors by decreased growth, enhanced dendricity, and melanogenesis. The rise in melanogenesis induced by NO-generating compounds is associated with an increased amount of both tyrosinase and tyrosinase-related protein 1. These observations suggest that NO plays an important role in the paracrine mediation of UV-induced melanogenesis.


Subject(s)
Keratinocytes/metabolism , Keratinocytes/radiation effects , Melanins/biosynthesis , Nitric Oxide/biosynthesis , Cell Division/drug effects , Cell Size/drug effects , Cells, Cultured , Culture Media, Conditioned , Dendrites , Humans , Keratinocytes/enzymology , Melanins/metabolism , Melanocytes/cytology , Melanocytes/drug effects , Nitric Oxide/physiology , Nitric Oxide/radiation effects , Nitric Oxide Synthase/chemistry , Ultraviolet Rays
11.
J Biol Chem ; 271(45): 28052-6, 1996 Nov 08.
Article in English | MEDLINE | ID: mdl-8910416

ABSTRACT

Ultraviolet B (UVB) radiation is the main physiological stimulus for human skin pigmentation; however, the molecular mechanisms underlying this process are still unclear. Recently, nitric oxide (NO) and cGMP have been involved in mediation of skin erythema induced by UVB. Therefore, we investigated the role of NO and cGMP in UVB-induced melanogenesis. In this study, we demonstrated that UVB stimulation of melanogenesis was mimicked by exogenous NO donors. Additionally, we showed that NO stimulated cGMP synthesis and that cGMP was also a potent stimulator of melanogenesis. Furthermore, the inhibition of the melanogenic effect of NO by guanylate cyclase inhibitor demonstrated that NO mediated its effect through the activation of guanylyl cyclase. Interestingly, 1 min after UVB irradiation, we observed a significant increase in cGMP content in melanocytes. The effects of UVB on cGMP production and on melanogenesis were blocked by both guanylate cyclase and NO synthase inhibitors. Additionally, inhibition of cGMP-dependent kinase also prevented the stimulation of melanogenesis by UVB and NO. Therefore, we concluded that NO and cGMP production is required for UVB-induced melanogenesis and that cGMP mediated its melanogenic effects mainly through the activation of cGMP-dependent kinase.


Subject(s)
Cyclic GMP/metabolism , Melanins/biosynthesis , Melanocytes/radiation effects , Nitric Oxide/metabolism , Signal Transduction , Ultraviolet Rays , Aminoquinolines/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Melanocytes/drug effects , Melanocytes/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine
12.
J Cell Sci ; 107 ( Pt 4): 1095-103, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8056833

ABSTRACT

Retinoic acid (RA) is a hormone-like agent involved in the control of cell differentiation. The most characteristic feature of melanocyte differentiation, melanogenesis, is stimulated by UV radiations. Excessive chronic sun exposure results in irregular skin hypermelanosis that can be partially corrected by topical RA. The basic mechanisms underlying this effect of RA are unknown. To determine whether RA can directly modulate excessive melanin synthesis, we analyzed the in vitro effect of cis- and trans-RA on UVB-induced melanogenesis in S91 mouse melanoma cells and in normal human melanocytes (NHM). In both cells types, the two RA isoforms significantly decreased the UVB-stimulated melanogenesis in term of tyrosinase activity and melanin neosynthesis. To correlate changes in melanogenesis with the expression of melanogenic enzymes, we determined the neosynthesis rate of tyrosinase, tyrosinase-related protein-1 (TRP-1/gp 75) and tyrosinase-related protein-2 (TRP-2/DOPAchrome tautomerase). Here we show that UVB-induced melanogenesis in NHM is related to an increased synthesis of tyrosinase and TRP-1 and to a dramatic decrease of TRP-2 expression. RA inhibition of UVB-induced melanogenesis acts at the post-transcriptional level leading to a decreased tyrosinase and TRP-1 synthesis. We also show that in NHM, inhibition of TRP-2 following UVB-treatment is significantly reversed by RA. This demonstrates a negative correlation between melanogenesis and TRP-2 expression.


Subject(s)
Intramolecular Oxidoreductases , Isomerases/biosynthesis , Melanins/biosynthesis , Melanocytes/drug effects , Melanocytes/radiation effects , Membrane Glycoproteins , Monophenol Monooxygenase/biosynthesis , Oxidoreductases , Protein Biosynthesis , Tretinoin/pharmacology , Ultraviolet Rays , Animals , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Enzyme Induction/drug effects , Enzyme Induction/radiation effects , Humans , Melanocytes/enzymology , Melanoma, Experimental/pathology , Mice , Skin/cytology
13.
J Cell Sci ; 106 ( Pt 4): 1015-22, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7907336

ABSTRACT

The major stimulus for human melanin production is ultraviolet (UV) radiation. Little is known about the mechanisms underlying this response and the eventual enzyme regulation resulting from this activation. We treated normal human melanocytes in culture with daily UVB radiations. Cumulative increases in UVB doses resulted in proportional increases in tyrosinase activity over the first few days whereas an intermittent pattern of tyrosinase activation was observed after the fifth day of irradiation. This intermittent pattern consisted of latency periods where no melanogenic response was elicited despite exposure to UVB. Tyrosinase activity in cellular extracts increased shortly after an effective irradiation, peaked at 3 hours and thereafter decreased to below basal levels. Increased tyrosinase activity was associated with increased amounts of both the newly synthesized and mature forms of the enzyme. Decreased tyrosinase activity following an activation period was correlated with decreases in both the expression of tyrosinase mRNA and the amount of the newly synthesized form of the enzyme present in the melanocytes 24 hours after six irradiations. This particular pattern of stimulation of tyrosinase was not observed in S-91 murine melanoma cells after repeated UVB irradiations. Taken together these results may suggest a photo-protective mechanism developed by irradiated normal human melanocytes.


Subject(s)
Melanins/biosynthesis , Melanocytes/radiation effects , Ultraviolet Rays , Animals , Cells, Cultured , Child , Dihydroxyphenylalanine/metabolism , Dose-Response Relationship, Radiation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Melanocytes/enzymology , Melanoma, Experimental/enzymology , Mice , Monophenol Monooxygenase/analysis , Periodicity , Skin/cytology , Tyrosine 3-Monooxygenase/analysis , White People
14.
Biol Reprod ; 46(5): 786-92, 1992 May.
Article in English | MEDLINE | ID: mdl-1317223

ABSTRACT

In vitro luteinization of bovine granulosa (LGC) and theca (LTC) cells was achieved by culturing cells with forskolin (10 microM) and insulin (2 micrograms/ml) for 9 days. This treatment induced the presence of cytochrome P450scc and adrenodoxin in both cell types, but to substantially higher levels in LGC than in LTC. Forskolin dose-dependently stimulated the secretion of progesterone and cAMP after 3 h of incubation in both cell types although LGC were less sensitive to this stimulation than were LTC. Only LTC were responsive to LH, in accordance with their higher LH/hCG binding capacity. Both prostaglandin F2 alpha (PGF2 alpha) and phorbol 12-myristate 13-acetate (TPA) increased progesterone production during 3 h incubation of LGC and LTC, and treatment with staurosporine (a protein kinase C inhibitor) reversed this effect. Neither TPA nor PGF2 alpha alone affected cAMP levels but each acted synergistically with forskolin to increase cAMP accumulation. These results indicate that 1) elevated progesterone output from LGC is related to steroidogenic enzyme level; 2) bovine LH (up to 100 ng/ml) does not provoke a response in LGC due to their low LH/hCG binding capacity; 3) cAMP-protein kinase A and protein kinase C pathways are both involved in progesterone production by LGC and LTC, possibly by enhancing cholesterol transport.


Subject(s)
Granulosa Cells/metabolism , Steroids/biosynthesis , Theca Cells/metabolism , Animals , Cattle , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/biosynthesis , Dinoprost/pharmacology , Female , Granulosa Cells/drug effects , Insulin/pharmacology , Luteinizing Hormone/metabolism , Luteinizing Hormone/pharmacology , Progesterone/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Theca Cells/drug effects
15.
Biol Reprod ; 43(6): 913-21, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2291928

ABSTRACT

This study was undertaken to investigate whether bovine granulosa and theca interna cells could be luteinized in vitro into luteal-like cells. Granulosa and theca cells were cultured for 9 days in the presence of forskolin (10 microM), insulin (2 micrograms/ml), insulin-like growth factor I (100 ng/ml), or a combination of these agents. During the first day of culture, granulosa and theca cells secreted estradiol and androstenedione, respectively; progesterone rose only after 3-5 days in culture and reached a maximum on the ninth day of culture. Cells incubated in the presence of forskolin plus insulin exhibited morphological and functional characteristics of luteal cells isolated from the corpus luteum. It was found that cell diameter, basal and stimulated progesterone secretion, and pattern of cell replication for both cell types were comparable to those of luteal cells. Numerous lipid droplets and intensified mitochondrial adrenodoxin staining also indicated active steroidogenesis in luteinized cells. After 9 days in culture, stimulants were withdrawn, and the culture proceeded in basal medium for an additional 5 days; elevated progesterone levels were maintained by luteinized granulosa cells (LGC), whereas in contrast a dramatic drop in progesterone production was observed in luteinized theca cells (LTC). On Day 9, cells were challenged for 3 h with LH (10 ng/ml), forskolin (10 microM), or cholera toxin (100 ng/ml), resulting in a 4-fold increase in progesterone secretion by LTC; the same treatments failed to stimulate progesterone in LGC.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Granulosa Cells/cytology , Theca Cells/cytology , Adrenodoxin/metabolism , Androstenedione/biosynthesis , Animals , Cattle , Cell Differentiation/drug effects , Cell Division , Colforsin/pharmacology , Estradiol/biosynthesis , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , In Vitro Techniques , Insulin/pharmacology , Luteal Cells/cytology , Luteal Cells/drug effects , Luteal Cells/metabolism , Luteinizing Hormone/pharmacology , Progesterone/biosynthesis , Theca Cells/drug effects , Theca Cells/metabolism
17.
Biol Reprod ; 36(3): 530-5, 1987 Apr.
Article in English | MEDLINE | ID: mdl-2885039

ABSTRACT

Rat oocytes resume meiosis spontaneously in vitro within 3 h after their isolation from the ovarian follicles. We report here that the spontaneous maturation of isolated rat oocytes is preceded by a drop in intracellular levels of cyclic adenosine 3',5'-monophosphate (cAMP). Further experiments were carried out to examine the possible correlation between the meiotic status and cAMP levels within the oocyte. To challenge rat cumulus-free oocytes to generate cAMP, bypassing their own adenylate cyclase, a preparation of an invasive adenylate cyclase from Bordetella pertussis was used. We found a dose-dependent elevation of cAMP levels within these oocytes that corresponded to inhibition of their spontaneous maturation. Persistent inhibition of meiosis was obtained with the continuous presence of the enzymatic preparation, whereas its removal resulted in a transient inhibition associated with a drop in cAMP. We suggest that the presence of elevated cAMP levels in the oocyte is directly responsible for the maintenance of meiotic arrest.


Subject(s)
Adenylyl Cyclases/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Animals , Bordetella pertussis/enzymology , Cyclic AMP/metabolism , Female , In Vitro Techniques , Oocytes/cytology , Oocytes/metabolism , Oogenesis/drug effects , Rats , Rats, Inbred Strains
18.
Biochem Biophys Res Commun ; 132(2): 570-4, 1985 Oct 30.
Article in English | MEDLINE | ID: mdl-2998374

ABSTRACT

Agonistic analogs of gonadotropin releasing hormone can induce oocyte maturation in rat follicle-enclosed oocytes (1-5). Cyclic AMP does not rise following exposure of the ovarian follicle to GnRH (3) suggesting that cAMP-dependent protein kinase is not involved in the mechanism of GnRH action in this system. Protein kinase C, which is independent of cAMP, has recently been reported to mediate GnRH action in the pituitary (6-8). The possible involvement of this enzyme in the regulation of oocyte maturation has been tested in the present study. We report here that phospholipase C and direct activators of protein kinase C can mimic the response of rat oocytes to GnRH. These results suggest that GnRH-induced meiotic maturation of rat oocytes is mediated by the phospholipid-dependent protein kinase, protein kinase C.


Subject(s)
Diglycerides/pharmacology , Glycerides/pharmacology , Oogenesis/drug effects , Phorbols/pharmacology , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cyclic AMP/analysis , Enzyme Activation/drug effects , Female , Gonadotropins, Equine/pharmacology , Meiosis/drug effects , Oocytes/drug effects , Oocytes/physiology , Protein Kinase C/physiology , Rats , Rats, Inbred Strains , Stimulation, Chemical
19.
Biol Reprod ; 31(2): 244-50, 1984 Sep.
Article in English | MEDLINE | ID: mdl-6089921

ABSTRACT

We have recently reported that the adenylate cyclase activator, forskolin, induces in the rat ovarian follicle both cAMP accumulation and oocyte maturation. We demonstrate here, on the other hand, that the spontaneous maturation in vitro of isolated rat cumulus-enclosed oocytes is inhibited by forskolin. The inhibitory effect of forskolin is dose dependent with an ED50 at 15 microM. Forskolin inhibition decreases gradually with time, being completely relieved by 20 h of culture. Methylisobutylxanthine significantly prolongs the duration of the inhibitory action of forskolin. In addition to its inhibitory effect on oocyte maturation, forskolin triggers the cumulus-oocyte complex to generate cAMP. Cyclic AMP accumulation is maximally stimulated by 100 microM of forskolin with an ED50 at 60 microM. The potency of the cumulus-oocyte complex to respond to forskolin in terms of cAMP accumulation decreases with time. The pattern of the decrease in the potency of the cumulus-oocyte complexes to generate cAMP corresponds with the relief of its inhibitory influence on the oocyte. These results indicate that inhibition of maturation of the cumulus-enclosed oocyte may be coupled to elevation of cAMP levels in the cumulus-oocyte complex. As isolated cumulus-free oocytes are not inhibited by forskolin, we suggest that in the cumulus-enclosed oocyte system, cAMP generated by the cumulus cells is apparently transferred to the oocyte and maintains it in a meiotically arrested state. Maturation in this system occurs upon relief of inhibition which results from cessation of cAMP generation by the cumulus cells.


Subject(s)
Diterpenes/pharmacology , Oocytes/drug effects , Oogenesis/drug effects , Ovarian Follicle/drug effects , Animals , Colforsin , Cyclic AMP/biosynthesis , Female , In Vitro Techniques , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...