Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Endocrinology ; 137(12): 5544-51, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8940382

ABSTRACT

In adrenal zona glomerulosa cells, the action of angiotensin II (Ang II) and of potassium (K+) on aldosterone synthesis is mediated by the Ca2+ messenger system. The major part of the steroidogenic pathway takes place inside the mitochondria, and Ca2+ must enter the mitochondrial matrix to stimulate the steroidogenic cascade. To examine how changes in the cytosolic free calcium concentration ([Ca2+]c) induced by Ang II and K+ are relayed into the mitochondrial matrix, we transfected bovine adrenal zona glomerulosa cells in primary culture with a chimeric complementary DNA encoding for the signal presequence targeting human cytochrome c oxidase subunit VIII to the matrix, linked to a complementary DNA coding for the Ca2+-sensitive photoprotein aequorin. Resting mitochondrial free calcium concentration ([Ca2+]m) amounted to 0.41 +/- 0.18 microM (n = 40). Ang II induced a concentration-dependent (EC50 = 11.3 +/- 6.0 nM), biphasic rise of [Ca2+]m. After a large transient initial peak (5.13 +/- 0.89 microM, n = 28), [Ca2+]m decreased to a plateau that remained higher than basal [Ca2+]m for several minutes in the presence of the hormone. By contrast, studies in cells transfected with cytosolic aequorin indicated that the rise of [Ca2+]c triggered by Ang II was confined to 1.34 +/- 0.26 microM (n = 17). In Ca2+-free medium, a reduced peak [Ca2+]m response to Ang II occurred without a secondary plateau. On readdition of extracellular Ca2+, in the presence of the hormone, the resulting Ca2+ influx was accompanied by small rise of [Ca2+]m. The mitochondrial uncoupler, carbonyl cyanide p-(trifluoro-methoxy)phenyl-hydrazone, prevented the Ang II-induced [Ca2+]m rise but not the [Ca2+]c response, thus demonstrating the mitochondrial location of transfected aequorin. In contrast to Ang II, K+ (13 mM) induced a sustained [Ca2+]c response, which was relayed without amplification into the mitochondrial matrix as a plateau of[Ca2+]m. This plateau of[Ca2+]m was suppressed by the addition of the dihydropyridine, nifedipine (200 nM). The inhibitor of the mitochondrial Na+/Ca2+ exchanger, CGP37157, reduced significantly the rate of decrease of [Ca2+]m following the peak induced by Ang II. In cells whose [Ca2+]c was clamped at various levels (0.05-0.860 microM) with ionomycin, a concentration-dependent stimulation of pregnenolone output was induced by Ca2+. Under these conditions, the output of pregnenolone--the early product of steroidogenesis--was markedly potentiated by CGP37157. These results suggest the existence of microdomains of high [Ca2+]c elicited by Ang II in the proximity of mitochondria. Moreover, our observations are consistent with a mitochondrial site of action for calcium in the activation of the steroidogenic cascade.


Subject(s)
Angiotensin II/pharmacology , Calcium/physiology , Mitochondria/metabolism , Potassium/pharmacology , Steroids/biosynthesis , Zona Glomerulosa/metabolism , Aequorin/genetics , Aequorin/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Cattle , Cells, Cultured , Cytosol/metabolism , Humans , Pregnenolone/biosynthesis , Sodium-Calcium Exchanger , Transfection , Zona Glomerulosa/cytology
2.
J Biol Chem ; 271(16): 9193-6, 1996 Apr 19.
Article in English | MEDLINE | ID: mdl-8621575

ABSTRACT

We have investigated the role of the essential Rho1 GTPase in cell integrity signaling in budding yeast. Conditional rho1 mutants display a cell lysis defect that is similar to that of mutants in the cell integrity signaling pathway mediated by protein kinase C (Pkc1), which is suppressed by overexpression of Pkc1.rho1 mutants are also impaired in pathway activation in response to growth at elevated temperature. Pkc1 co-immunoprecipitates with Rho1 in yeast extracts, and recombinant Rho1 associates with Pkc1 in vitro in a GTP-dependent manner. Recombinant Rho1 confers upon Pkc1 the ability to be stimulated by phosphatidylserine, indicating that Rho1 controls signal transmission through Pkc1.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Saccharomyces cerevisiae/enzymology , rho GTP-Binding Proteins , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Enzyme Activation , GTP Phosphohydrolases/isolation & purification , GTP-Binding Proteins/isolation & purification , Guanosine Triphosphate/metabolism , Models, Biological , Mutagenesis, Site-Directed , Protein Kinase C/isolation & purification , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins , Temperature , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae
3.
Science ; 272(5259): 279-81, 1996 Apr 12.
Article in English | MEDLINE | ID: mdl-8602515

ABSTRACT

1,3-beta-D-glucan synthase [also known as beta(1-->3) glucan synthase] is a multi-enzyme complex that catalyzes the synthesis of 1,3-beta-linked glucan, a major structural component of the yeast cell wall. Temperature-sensitive mutants in the essential Rho-type guanosine triphosphatase (GTPase), Rho1p, displayed thermolabile glucan synthase activity, which was restored by the addition of recombinant Rho1p. Glucan synthase from mutants expressing constitutively active Rho1p did not require exogenous guanosine triphosphate for activity. Rho1p copurified with beta(1-->3)glucan synthase and associated with the Fks1p subunit of this complex in vivo. Both proteins were localized predominantly at sites of cell wall remodeling. Therefore, it appears that Rho1p is a regulatory subunit of beta(1-->3)glucan synthase.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Glucosyltransferases/metabolism , Membrane Proteins , Multienzyme Complexes/metabolism , Saccharomyces cerevisiae/enzymology , Schizosaccharomyces pombe Proteins , rho GTP-Binding Proteins , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/isolation & purification , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , GTP-Binding Proteins/isolation & purification , Glucosyltransferases/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , Multienzyme Complexes/chemistry , Protein Kinase C/metabolism , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins , Temperature
4.
J Biol Chem ; 270(25): 15137-42, 1995 Jun 23.
Article in English | MEDLINE | ID: mdl-7797497

ABSTRACT

In adrenal glomerulosa cells, low threshold voltage-activated (T-type) calcium channels play a crucial role in coupling physiological variations of extracellular potassium to aldosterone biosynthesis. Angiotensin II markedly reduced the activity of these channels by shifting their activation curve toward positive voltage values. This inhibition of the channels resulted in a marked decrease of the cytosolic free calcium concentration maintained by potassium. This effect was abolished by losartan, a specific antagonist of the angiotensin II AT1 receptor. Hormone action on T-type channels appeared to be mediated by protein kinase C because 1) it was mimicked by phorbol ester and diacylglycerol, and 2) it was significantly reduced by decreasing protein kinase C activity with specific inhibitors such as chelerythrine chloride or a pseudosubstrate of the enzyme, as well as by protein kinase C down-regulation. Similarly, protein kinase C activation reduced the cytosolic calcium response to potassium and the steroidogenic action of this agonist. Low threshold T-type calcium channels therefore appear as potential sites for the modulation of steroidogenesis by protein kinase C in adrenal glomerulosa cells.


Subject(s)
Angiotensin II/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Protein Kinase C/metabolism , Zona Glomerulosa/physiology , Aldosterone/metabolism , Animals , Calcium/metabolism , Cattle , Cells, Cultured , Cytosol/metabolism , Diglycerides/pharmacology , Drug Interactions , Enzyme Activation , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Zona Glomerulosa/drug effects
5.
Biochem J ; 305 ( Pt 2): 569-76, 1995 Jan 15.
Article in English | MEDLINE | ID: mdl-7832775

ABSTRACT

The Ca(2+)-messenger system plays a crucial role in the regulation of steroid production in adrenal zona-glomerulosa cells, as it is known to mediate the action of both angiotensin II and K+. In the present study we used intact isolated glomerulosa cells in which the cytosolic free Ca2+ concentration ([Ca2+]c) was clamped at various levels with the Ca2+ ionophore ionomycin in order to locate the site(s) of action of Ca2+. By measuring in parallel steroid synthesis and [Ca2+]c, we show that Ca2+ levels (50-860 nM) regulate the production of both pregnenolone (up to 669 +/- 71.1% of the basal production) and aldosterone (up to 301 +/- 42.2%; EC50 = 303 nM). By contrast, Ca2+ did not stimulate the conversion of 11-deoxycorticosterone into aldosterone. Ca2+ modulation did not affect the formation of pregnenolone from freely diffusible analogues of cholesterol, indicating that Ca2+ acts at a step upstream of cholesterol side-chain cleavage. Moreover cycloheximide, an inhibitor of protein translation and of adrenocorticotropin-induced facilitation of intramitochondrial cholesterol transport, the rate-limiting step in steroidogenesis, also blocked Ca(2+)-triggered pregnenolone formation. This is consistent with a model in which Ca2+ promotes cholesterol transfer between mitochondrial membranes. In addition, agents using the cyclic AMP pathway as well as angiotensin II potentiated the steroidogenic response to increases in [Ca2+]c by augmenting both the efficacy and the potency of Ca2+. This effect of angiotensin II did not involve protein kinase C. These results establish a direct link between agonist-induced [Ca2+]c rises and a specific step of the steroidogenic pathway.


Subject(s)
Aldosterone/biosynthesis , Calcium/pharmacology , Pregnanolone/biosynthesis , Zona Glomerulosa/metabolism , Angiotensin II/pharmacology , Animals , Bucladesine/pharmacology , Cattle , Cholesterol/metabolism , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Hydroxycholesterols/pharmacology , Ionomycin/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Zona Glomerulosa/cytology , Zona Glomerulosa/drug effects
6.
Endocrinology ; 135(2): 751-8, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8033823

ABSTRACT

Angiotensin-II (AngII)-induced Ca2+ influx in adrenal glomerulosa cells, a signal necessary for the stimulation of steroidogenesis by the hormone, is believed to involve two distinct mechanisms: 1) opening of voltage-operated Ca2+ channels, and 2) activation of a capacitative Ca2+ entry pathway that is dependent on calcium release from intracellular stores. Nicardipine, a dihydropyridine calcium antagonist, has been used to investigate the role of these Ca2+ entry mechanisms in the steroidogenic response to AngII. As demonstrated with the patch-clamp technique, micromolar concentrations of nicardipine completely blocked voltage-operated Ca2+ channel activity of both T- and L-types. This agent similarly inhibited the rise of cytosolic free calcium concentration induced by potassium, but did not significantly affect the response to thapsigargin, an activator of the capacitative pathway. Nicardipine reduced by only 22% the calcium influx stimulated by AngII, and the nicardipine-insensitive part of this response was abolished after exhausting the intracellular Ca2+ stores with thapsigargin. Similarly, aldosterone secretion induced by AngII was only partially inhibited (40%) by nicardipine at concentrations that completely abolished the steroidogenic response to potassium. Thapsigargin by itself was able to stimulate aldosterone production, an action highly potentiated by physiological concentrations of extracellular potassium. These data strongly suggest that the major part of the calcium influx response to AngII, leading to aldosterone formation, involves a capacitative calcium entry pathway activated by the release of calcium from intracellular stores. This mechanism of calcium influx could be responsible for some features of aldosterone response to the hormone, such as its poor sensitivity to dihydropyridines or its potentiation by potassium.


Subject(s)
Aldosterone/biosynthesis , Angiotensin II/pharmacology , Calcium/metabolism , Zona Glomerulosa/metabolism , Aldosterone/metabolism , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , Cattle , Cells, Cultured , Nicardipine/pharmacology , Potassium/pharmacology , Terpenes/pharmacology , Thapsigargin , Zona Glomerulosa/drug effects
7.
Biochem J ; 296 ( Pt 2): 309-12, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8257418

ABSTRACT

Thapsigargin, an inhibitor of the microsomal Ca2+ pumps, has been extensively used to study the intracellular Ca2+ pool participating in the generation of the agonist-induced Ca2+ signal in various cell types. A dual effect of this agent was observed in bovine adrenal zona glomerulosa cells. At nanomolar concentrations, thapsigargin stimulated a sustained Ca2+ influx, probably resulting from Ca(2+)-store depletion. In contrast, when added at micromolar concentrations, thapsigargin prevented the rise in cytosolic free Ca2+ concentration ([Ca2+]c) induced by K+. This inhibitory effect of thapsigargin on voltage-activated Ca2+ channels was confirmed by measuring Ba2+ currents by the patch-clamp technique. Both low-threshold (T-type) and high-threshold (L-type) Ca2+ channels were affected by micromolar concentrations of thapsigargin. Analysis of the current-voltage relationship for T-type channels revealed that thapsigargin did not modify the sensitivity of these channels to the voltage, but decreased the maximal current flowing through the channels. In conclusion, thapsigargin appears to exert a dual effect on adrenal glomerulosa cells. At lower concentrations, this agent induces a sustained Ca2+ entry, whereas at higher concentrations it decreases [Ca2+]c by blocking voltage-activated Ca2+ channels.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium/metabolism , Terpenes/pharmacology , Zona Glomerulosa/physiology , Animals , Barium/pharmacology , Cattle , Cells, Cultured , Electric Stimulation , Kinetics , Membrane Potentials/drug effects , Potassium/pharmacology , Thapsigargin , Zona Glomerulosa/drug effects
8.
Endocrinology ; 133(3): 1213-20, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8365363

ABSTRACT

The homeostasis of cytosolic free calcium ([Ca2+]i) and intracellular free sodium ([Na+]i) are linked in many cell types. We, therefore, studied the effect on [Na+]i of two physiological stimulators of aldosterone synthesis that trigger the calcium messenger system, angiotensin-II (Ang II) and potassium ion (K+), in cultured bovine adrenal glomerulosa cells, using the intracellular fluorescent probe for sodium, sodium benzofuran isophthalate. Ang II induced a concentration-dependent and sustained increase in [Na+]i, from a resting value of 9.2 +/- 3.5 to a maximum of 48.5 +/- 5.5 mM (n = 14). This [Na+]i response was mediated by receptors of the AT1 subtype, because it was abolished by losartan (DuP 753). K+ (15 mM) induced a weaker [Na+]i response, from 5.9 +/- 2.6 to 16.8 +/- 2.5 mM (n = 9). In freshly prepared cells, basal [Na+]i was significantly higher (23.9 +/- 1.8 mM; n = 14; P < 0.01) than in cultured cells. Atrial natriuretic peptide, which is known to affect sodium transport in various cell types, did not alter the [Na+]i response elicited by Ang II. Ethylisopropylamiloride, an inhibitor of Na+/H+ exchange, and dichlorobenzamyl, an inhibitor of Na+/Ca2+ exchange, both inhibited in a concentration-dependent manner the Ang II- and K(+)-induced aldosterone response. Isoosmotic replacement of extracellular Na+ markedly reduced basal aldosterone synthesis. Under these conditions, the concentration-response curve for Ang II-induced aldosterone synthesis was shifted to the right, and its maximum was strikingly diminished. These results show that Ang II and, to a lesser extent, K+ induce significant changes in [Na+]i in bovine glomerulosa cells. These [Na+]i changes probably occur through the Na+/H+ and Na+/Ca2+ exchangers and are likely to play a role in activation of the steroidogenic cascade.


Subject(s)
Aldosterone/biosynthesis , Angiotensin II/pharmacology , Sodium/metabolism , Zona Glomerulosa/metabolism , Animals , Atrial Natriuretic Factor/pharmacology , Benzofurans , Biological Transport , Carrier Proteins/metabolism , Cattle , Cells, Cultured , Cytosol/metabolism , Ethers, Cyclic , Fluorescent Dyes , Homeostasis , Potassium/pharmacology , Sodium-Calcium Exchanger , Zona Glomerulosa/drug effects
9.
Endocrinology ; 132(4): 1489-96, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8384990

ABSTRACT

Angiotensin-II (Ang-II), K+, and ACTH are important stimulators of aldosterone secretion that require Ca2+ influx to be active. However, Ang-II and K+ are linked to the Ca2+ messenger system, while ACTH is coupled to the cAMP pathway. Peripheral-type binding sites for benzodiazepines are particularly abundant in steroidogenic tissues and have been proposed to be involved in the steroidogenic action of ACTH in Y-1 adrenocortical cells. We report here that in adrenal glomerulosa cells, peripheral-type [4'-chlor-diazepam (CDZ), 1-(2-chlorophenyl)N-methyl-N-(1-methylpropyl)3-isoquinolinecarboxamid e (RP 52028), and flunitrazepam], but not a central-type (flumazenil) benzodiazepine reversibly abolished the stimulation of aldosterone output induced by Ang-II or K+, while they had no significant effect on basal aldosterone secretion. This inhibitory effect depended upon drug concentration (IC50 30 microM for CDZ) and affected the potencies of both stimulators, without altering their respective EC50 values. Similar results were obtained when aldosterone production was stimulated with ACTH, forskolin, or (Bu)2cAMP. Aldosterone production from exogenous 25-hydroxycholesterol or progesterone was partially inhibited by CDZ. In glomerulosa cells loaded with a fluorescent Ca2+ probe, benzodiazepines blocked Ca2+ influx triggered by K+ or Ang-II without affecting the release of Ca2+ from intracellular stores induced by Ang-II. T- and L-type Ca2+ channel activities, monitored with the patch-clamp technique, were both inhibited within the same range of concentrations as aldosterone synthesis and Ca2+ influx. These results indicate that in adrenal zona glomerulosa cells, peripheral-type benzodiazepines block Ca2+ influx through voltage-activated channels. The combined action of peripheral-type benzodiazepines on calcium influx and precursor conversion may be responsible for the observed inhibition of Ang-II-, K(+)-, or ACTH-induced aldosterone secretion.


Subject(s)
Aldosterone/biosynthesis , Benzodiazepines/pharmacology , Calcium Channel Blockers/pharmacology , Zona Glomerulosa/metabolism , Animals , Barium/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Osmolar Concentration , Zona Glomerulosa/cytology
10.
Endocrinology ; 132(3): 1035-43, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8382595

ABSTRACT

Tetrandrine, an alkaloid extracted from a Chinese medicinal herb traditionally used in hypertension treatment, inhibited aldosterone production induced in bovine adrenal glomerulosa cells by either potassium ion, angiotensin II, or ACTH in a concentration-dependent manner (IC50 = 10 microM). The inhibition of the response to potassium by tetrandrine had a pattern very similar to that of nickel, a blocker of T-type calcium channels. In addition, tetrandrine prevented calcium influx induced by potassium or angiotensin II without affecting the calcium release phase stimulated by the hormone. The effect of tetrandrine on voltage-activated barium currents was investigated using the whole cell configuration of the patch clamp technique. T-type currents were isolated by recording the slowly deactivating currents elicited during repolarization of the cell to -65 mV after various depolarizing pulses. These currents were blocked by micromolar concentrations of the drug. The voltage sensitivity of channel activation was not affected by tetrandrine; nevertheless, the drug significantly slowed the deactivation of the current. The action of tetrandrine did not require the activation of the channel. Tetrandrine also affected L-type currents, as assessed after inactivating T channels for 100 msec, but at higher concentrations of the drug. Thus, tetrandrine affects with a similar potency aldosterone production, calcium influx, and T-type calcium channel activity. This finding strongly suggests a role for these channels in calcium signaling and control of steroidogenesis in adrenal glomerulosa cells.


Subject(s)
Aldosterone/biosynthesis , Alkaloids/pharmacology , Benzylisoquinolines , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium/metabolism , Mineralocorticoid Receptor Antagonists/pharmacology , Zona Glomerulosa/metabolism , Adrenocorticotropic Hormone/pharmacology , Angiotensin II/pharmacology , Animals , Cadmium/pharmacology , Calcium Channels/drug effects , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Fura-2 , Kinetics , Membrane Potentials/drug effects , Nickel/pharmacology , Potassium Chloride/pharmacology , Zona Glomerulosa/drug effects , Zona Glomerulosa/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...