Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Cell Endocrinol ; 264(1-2): 90-101, 2007 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-17092635

RESUMO

Stanniocalcin-1 (STC-1) is one of only a handful of hormones that are targeted to mitochondria. High affinity receptors for STC-1 are present on cytoplasmic membranes and both the outer and inner mitochondrial membranes of nephron cells and hepatocytes. In both cell types, STC-1 is also present within the mitochondrial matrix and receptors presumably enable its sequestration. Furthermore, studies in bovine heart sub-mitochondrial particles have shown that STC-1 has concentration-dependent stimulatory effects on electron transport chain activity. The aim of the present study was to determine if the same effects could be demonstrated in intact, respiring mitochondria. At the same time, we also sought to demonstrate the functionality, if any, of an ATP binding cassette that has only recently been identified within the N-terminus of STC-1 by Prosite analysis. Intact, respiring mitochondria were isolated from rat muscle and liver and exposed to increasing concentrations of recombinant human STC-1 (STC-1). Following a 1h exposure to 500 nM STC-1, mitochondria from both organs displayed significant increases in respiration rate as compared to controls. Moreover, STC-1 uncoupled oxidative phosphorylation as ADP:O ratios were significantly reduced in mitochondria from both tissues. The resulting uncoupling was correlated with enhanced mitochondrial (45)Ca uptake in the presence of hormone. Respiratory studies were also conducted on a mouse inner medullary collecting cell line, where STC-1 had time and concentration-dependent stimulatory effects within the physiological range. In the presence of nucleotide triphosphates such as ATP and GTP (5mM) the respiratory effects of STC-1 were attenuated or abolished. Receptor binding studies revealed that this was due to a four-fold decrease in binding affinity (KD) between ligand and receptor. The results suggest that STC-1 stimulates mitochondrial electron transport chain activity and calcium transport, and that these effects are negatively modulated by nucleotide triphosphates.


Assuntos
Trifosfato de Adenosina/farmacologia , Glicoproteínas/farmacologia , Guanosina Trifosfato/farmacologia , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Musculares/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Animais , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Glicoproteínas/metabolismo , Masculino , Ratos , Ratos Wistar
2.
Mol Cell Endocrinol ; 186(1): 111-9, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11850127

RESUMO

The secretion of parathyroid hormone (PTH) and calcitonin (CT) in mammals are both tightly regulated by the prevailing levels of extracellular ionic calcium (Ca(2+)). And, it is now widely recognized that both of these Ca(2+) effects are mediated exclusively through a seven transmembrane calcium sensing receptor or CaR. As in the case of PTH and CT, the secretion of stanniocalcin (STC) in fish is tightly regulated by the levels of extracellular Ca(2+). Fish STC functions as an anti-hypercalcemic hormone such that a rise in extracellular Ca(2+) above the physiological set-point of approximately 1.2 mM provokes an immediate secretory response. Whether or not Ca(2+)-regulated STC secretion in fishes is mediated by similar type of receptor has never been addressed. Here, we have found that Ca(2+)-stimulated STC secretion in salmon is mimicked by CaR mimetics, pharmacological agents that increase the sensitivity of the CaR to calcium. NPS 467, a small organic molecule that acts as a positive allosteric modulator of the CaR and alters calciotropic hormone secretion in mammals, was examined for effects on serum levels of STC in trout. The IP administration of NPS R-467 had time and dose-dependent stimulatory effects on STC secretion that were indistinguishable from those of Ca(2+) loading. The effects of NPS 467 were stereospecific and had no effects on serum CT. NPS 467 induced STC release was also manifested by a downstream physiological response; the inhibition of gill calcium transport. A cDNA clone was amplified from a fish corpuscle of Stannius cDNA library with high homology to the human CaR. RT-PCR revealed that this transcript was also present in gill, kidney, pancreas, brain, muscle and spleen. These findings suggest that Ca(2+)-stimulated STC secretion in fishes is mediated by a calcium ion-sensing receptor similar to that in mammals.


Assuntos
Compostos de Anilina/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/agonistas , Glicoproteínas/metabolismo , Hormônios/metabolismo , Oncorhynchus mykiss/metabolismo , Animais , Transporte Biológico , Calcitonina/sangue , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Cátions Bivalentes , DNA Complementar/análise , Relação Dose-Resposta a Droga , Glândulas Endócrinas/efeitos dos fármacos , Glândulas Endócrinas/metabolismo , Glicoproteínas/sangue , Hormônios/sangue , Oncorhynchus mykiss/sangue , Reação em Cadeia da Polimerase
3.
Am J Physiol Renal Physiol ; 294(4): F788-94, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18199603

RESUMO

Kidney collecting duct principal cells are the main source of stanniocalcin-1 (STC-1) production and secretion. From there, the hormone targets thick ascending limb and distal convoluted tubule cells, as well as collecting duct cells. More specifically, STC-1 targets their mitochondria to exert putative antiapoptotic effects. Two distal tubule cell lines serve as models of STC-1 production and/or mechanism of action. Madin-Darby canine kidney-1 (MDCK-1) cells mimic collecting duct cells in their synthesis of STC-1 ligand and receptor, whereas inner medullary collecting duct-3 (IMCD-3) cells respond to additions of STC-1 by increasing their respiration rate. In the present study, MDCK cell STC-1 secretion was examined under normal and hypertonic conditions, vectorally, and in response to hormones and signal transduction pathway activators/inhibitors. STC-1 receptor regulation was monitored in both cell lines in response to changing ligand concentration. The results showed that NaCl-induced hypertonicity had concentration-dependent stimulatory effects on STC-1 secretion, as did the PKC activator TPA. Calcium and ionomycin were inhibitory, whereas calcium receptor agonists had no effect. Angiotensin II, aldosterone, atrial natriuretic factor, antidiuretic hormone, and forskolin also had no effects. Moreover, STC-1 secretion exhibited no vectoral preference. STC-1 receptors were insensitive to homologous downregulation in both cell lines. In contrast, they were upregulated when STC-1 secretion was inhibited by calcium. The findings suggest that hypertonicity-induced STC-1 secretion is regulated through PKC activation and that high intracellular calcium levels are a potent inhibitor of release. More intriguingly, the results suggest that the receptor may not accompany STC-1 in its passage to the mitochondria.


Assuntos
Glicoproteínas/genética , Receptores de Superfície Celular/genética , Animais , Cloreto de Cálcio/farmacologia , Linhagem Celular , Cães , Ativação Enzimática , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Rim , Proteína Quinase C/metabolismo , Cloreto de Sódio/farmacologia
4.
Am J Physiol Endocrinol Metab ; 291(5): E974-81, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16772321

RESUMO

During pregnancy and lactation in rodents, stanniocalcin-1 (STC-1) production by the ovaries is upregulated markedly and released into the circulation. The mammary glands are one target of this systemically delivered hormone. The purpose of this study was to lower serum levels of STC-1 in lactating mice through passive immunization so as to monitor the effects on mammary gland function and postnatal pup growth. Passive immunization significantly reduced circulating hormone levels, and pup growth was significantly compromised (30%), even though control and experimental litters had ingested equal amounts of milk. When mammary glands were analyzed, the alveolar area was significantly reduced in antibody-treated mothers. An analysis of milk composition revealed no changes in lactose, protein, or electrolyte levels but an approximately 40% reduction in triglyceride levels. The latter was due to a significant reduction in mammary gland lipoprotein lipase activity and led to a significant buildup of triglycerides in the serum. Body fat content was also significantly reduced in pups from antibody-treated mothers, whereas pup fecal fat content was increased. In mothers, passive immunization also caused significant behavioral effects, in particular, increased locomotor and hindleg rearing activities. Collectively, the results suggest that systemically derived STC-1 has important effects on mammary gland development and the transfer of serum-based triglycerides into milk. Locomotor effects suggest that STC-1 also has a role in maternal behavior.


Assuntos
Glicolipídeos/metabolismo , Glicoproteínas/sangue , Glicoproteínas/metabolismo , Lactação/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Tecido Adiposo/crescimento & desenvolvimento , Animais , Animais Lactentes , Desenvolvimento Ósseo , Fezes , Feminino , Glicoproteínas/imunologia , Imunização Passiva , Gotículas Lipídicas , Comportamento Materno/fisiologia , Camundongos , Camundongos Endogâmicos , Receptores de Superfície Celular/metabolismo , Triglicerídeos/sangue
5.
Can J Physiol Pharmacol ; 83(11): 953-6, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16391703

RESUMO

There are 2 forms of stanniocalcin (STC) produced by the STC-1 gene; a 50 kDa polypeptide known as STC50 and a recently discovered group of higher molecular weight variants that are collectively referred to as big STC. Both have different tissue patterns of expression and different intracellular targeting pathways. STC50 functions locally in tissues such as muscle, liver, and kidney and is targeted to mitochondria. Big STC, on the other hand, is made by the ovaries. It signals both locally on nearby corpus luteal cells and systemically. Interestingly, however, receptor binding assays employing STC50 as the tracer have shown that the smaller ligand can bind equally to tissue receptors targeted by either form of the hormone. This suggests there may be cross-talk between ligands. The present study provides credence to this notion by demonstrating how the 2 hormones can compete for tissue receptors normally targeted by 1 form of the hormone (big STC). The results also reveal how STC50 can completely block the inhibitory effects of big STC on luteal cell progesterone release when added simultaneously. The findings therefore add credence to the possibility that there may be circumstances during which the 2 ligands functionally antagonize each other's actions.


Assuntos
Corpo Lúteo/metabolismo , Glicoproteínas/metabolismo , Animais , Ligação Competitiva , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Corpo Lúteo/citologia , Feminino , Humanos , Microssomos/metabolismo , Progesterona/metabolismo
6.
Kidney Int ; 67(2): 477-82, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15673295

RESUMO

BACKGROUND: The 50 kD form of the hormone stanniocalcin-1 (STC50) is widely distributed in organs such as kidney, lung, and liver. Kidney collecting duct cells produce STC50 for local targeting to proximal tubule cells to increase phosphate reabsorption. As such the current dogma is that in most organs STC50 is a purely local mediator that is not released into the circulation. However, liver hepatocytes contain high levels of both STC50 and its receptor but little evidence of STC production, suggesting that the hormone may in fact be delivered to hepatocytes systemically. Moreover, previous data suggest that red blood cells may in fact bind STC. In this report, we have sought to identify STC binding activity in mammalian blood. METHODS: Human, pig, and dog red blood cells were analyzed in STC receptor binding assays. Mouse red blood cells and adult mouse kidney were also analyzed histologically for the presence of STC ligand and receptor. RESULTS: Saturable, high affinity STC receptors were identified on red blood cells from all species. More intriguingly, STC binding activity was also identified in glomerular filtrate, indicative of a soluble, filterable STC binding protein. This binding protein was subsequently observed being reabsorbed in proximal straight tubules. CONCLUSION: These findings suggest that our inability to detect STC in mammalian serum is due to its being attached to soluble and tethered forms of a high-affinity binding protein. This could be a means of delivering STC to distant targets as well as a mechanism for removing unwanted hormone from the circulation.


Assuntos
Eritrócitos/metabolismo , Glicoproteínas/metabolismo , Glomérulos Renais/metabolismo , Receptores de Superfície Celular/sangue , Animais , Cães , Humanos , Imuno-Histoquímica , Suínos
7.
Am J Physiol Endocrinol Metab ; 289(2): E197-205, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15741242

RESUMO

The hormone stanniocalcin (STC) is widely distributed, and in rodents the highest levels of expression are in the ovaries. In both cows and rodents, ovarian STC consists of three high-molecular-weight variants collectively known as big STC. In the ovary, big STC is made by theca cells and interstitial cells and is targeted to lipid storage droplets of nearby luteal cells to inhibit progesterone release. An endocrine pathway is operative during pregnancy and lactation. Whether or not big STC is made by tissues other than ovary has never been addressed. Therefore, the purpose of this study was to determine via a detailed characterization of adrenal glands and adipocytes whether big STC is present in other cells that store and release lipids. The results showed that STC was made in bovine and mouse adrenals, mainly in steroidogenic, adrenocortical cells. The majority of ligand and receptor were likewise confined to cortical zone cells. As in luteal cells, high levels of ligand and receptor were found in the adrenocortical cell lipid droplet fraction. However, adrenals made only the largest (135 kDa) of the three big STC variants. Nonetheless, adrenal STC had much greater receptor affinity than a mixture of the three big STC variants. Adipocytes contained all three big STC variants, and both ligand and receptor were heavily concentrated on the lipid droplets. Moreover, adipocyte lipid storage droplets had 50-fold more receptors than those in steroidogenic cells, indicating that big STC is heavily targeted to adipose cells. The findings collectively support the hypothesis that big STC is not unique to ovarian steroidogenic cells but is in fact common to cells with a role in lipid storage and release.


Assuntos
Adipócitos/metabolismo , Córtex Suprarrenal/metabolismo , Corpo Lúteo/metabolismo , Glicoproteínas/metabolismo , Adipócitos/citologia , Córtex Suprarrenal/citologia , Animais , Bovinos , Fracionamento Celular , Células Cultivadas , Corpo Lúteo/citologia , Feminino , Imuno-Histoquímica , Metabolismo dos Lipídeos , Isoformas de Proteínas
8.
Am J Physiol Endocrinol Metab ; 289(4): E634-42, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16150955

RESUMO

In most mammalian tissues, the stanniocalcin-1 gene (STC-1) produces a 50-kDa polypeptide hormone known as STC50. Within the ovaries, however, the STC-1 gene generates three higher-molecular-mass variants known as big STC. Big STC is targeted locally to corpus luteal cells to block progesterone release. During pregnancy and lactation, however, ovarian big STC production increases markedly, and the hormone is released into the serum. During lactation, this increase in hormone production is dependent on a suckling stimulus, suggesting that ovarian big STC may have regulatory effects on the lactating mammary gland. In this report, we have addressed this possibility. Our results revealed that virgin mammary tissue contained large numbers of membrane- and mitochondrial-associated STC receptors. However, as pregnancy progressed into lactation, there was a decline in receptor densities on both organelles and a corresponding rise in nuclear receptor density, most of which were on milk-producing, alveolar cells. This was accompanied by nuclear sequestration of the ligand. Sequestered STC resolved as one approximately 135-kDa band in the native state and therefore had the appearance of a big STC variant. However, chemical reduction collapsed this one band into six closely spaced, lower-molecular-mass species (28-41 kDa). Mammary gland STC production also underwent a dramatic shift during pregnancy and lactation. High levels of STC gene expression were observed in mammary tissue from virgin and pregnant rats. However, gene expression then fell to nearly undetectable levels during lactation, coinciding with the rise in nuclear targeting. These findings have thus shown that the mammary glands are indeed targeted by STC, even in the virgin state. They have further shown that there are marked changes in this targeting pathway during pregnancy and lactation, accompanied by a switch in ligand source (endogenous to exogenous). They also represent the first example of nuclear targeting by STC.


Assuntos
Núcleo Celular/metabolismo , Glicoproteínas/metabolismo , Lactação/metabolismo , Glândulas Mamárias Animais/metabolismo , Prenhez/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Gravidez , Distribuição Tecidual
9.
J Biol Chem ; 277(47): 45249-58, 2002 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-12223480

RESUMO

The polypeptide hormone stanniocalcin (STC) is widely expressed in mammalian tissues. STC acts locally in kidney and gut to modulate calcium and phosphate excretion, and its overexpression in mice results in high serum phosphate, dwarfism, and increased metabolic rate. STC has also been linked to cancer, pregnancy, lactation, angiogenesis, organogenesis, cerebral ischemia, and hypertonic stress. In this report we have characterized the STC receptor and the functional targeting of ligand and receptor to mitochondria. For receptor binding analysis, a stanniocalcin-alkaline phosphatase fusion protein was engineered. Subsequent binding assays using the fusion protein indicated that kidney and liver contained the highest number of binding sites with affinities of 0.8 and 0.25 nm, respectively. Intriguingly, purified mitochondria from both tissues yielded similar high affinity binding sites. Fractionation analysis revealed that the majority of binding sites were localized to the inner mitochondrial membrane. In further studies, we characterized the time course of STC-alkaline phosphatase fusion protein sequestration by intact mitochondria. In situ ligand binding also revealed discrete, displaceable binding to plasma membranes and mitochondria of nephron cells and liver hepatocytes. The existence of mitochondrial receptors prompted a similar search for the ligand. Immunogold electron microscopy revealed that STC was preferentially concentrated in the mitochondria of all nephron segments targeted by STC. Subcellular fractionation revealed that >90% of cellular STC immunoreactivity was mitochondrial, confined to the inner matrix, and similar in size to recombinant STC (50 kDa). In functional studies, recombinant STC had concentration-dependent stimulatory effects on electron transfer by sub-mitochondrial particles. Collectively the evidence implies a role for STC in cell metabolism.


Assuntos
Glicoproteínas/metabolismo , Hormônios/metabolismo , Mitocôndrias/metabolismo , Transporte Proteico/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Sítios de Ligação , Bovinos , Fracionamento Celular , Linhagem Celular , Membrana Celular/metabolismo , Cães , Transporte de Elétrons , Glicoproteínas/genética , Coração/anatomia & histologia , Coração/fisiologia , Hormônios/genética , Humanos , Rim/química , Rim/citologia , Rim/metabolismo , Ligantes , Fígado/química , Fígado/citologia , Fígado/metabolismo , Camundongos , Mitocôndrias/ultraestrutura , Mitocôndrias Hepáticas/metabolismo , Ligação Proteica , Ratos , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA