Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 5(7)2016 07 21.
Article in English | MEDLINE | ID: mdl-27444511

ABSTRACT

BACKGROUND: Tubulointerstitial fibrosis, the final outcome of most kidney diseases, involves activation of epithelial mesenchymal transition (EMT). Endothelin-1 (ET-1) activates EMT in cancer cells, but it is not known whether it drives EMT in the kidney. We therefore tested the hypothesis that tubulointerstitial fibrosis involves EMT driven by ET-1. METHODS AND RESULTS: Transgenic TG[mRen2]27 (TGRen2) rats developing fulminant angiotensin II-dependent hypertension with prominent cardiovascular and renal damage were submitted to drug treatments targeted to ET-1 and/or angiotensin II receptor or left untreated (controls). Expressional changes of E-cadherin and α-smooth muscle actin (αSMA) were examined as markers of renal EMT. In human kidney HK-2 proximal tubular cells expressing the ETB receptor subtype, the effects of ET-1 with or without ET-1 antagonists were also investigated. The occurrence of renal fibrosis was associated with EMT in control TGRen2 rats, as evidenced by decreased E-cadherin and increased αSMA expression. Irbesartan and the mixed ET-1 receptor antagonist bosentan prevented these changes in a blood pressure-independent fashion (P < 0.001 for both versus controls). In HK-2 cells ET-1 blunted E-cadherin expression, increased αSMA expression (both P < 0.01), collagen synthesis, and metalloproteinase activity (P < 0.005, all versus untreated cells). All changes were prevented by the selective ETB receptor antagonist BQ-788. Evidence for involvement of the Rho-kinase signaling pathway and dephosphorylation of Yes-associated protein in EMT was also found. CONCLUSIONS: In angiotensin II-dependent hypertension, ET-1 acting via ETB receptors and the Rho-kinase and Yes-associated protein induces EMT and thereby renal fibrosis.


Subject(s)
Actins/drug effects , Angiotensin Receptor Antagonists/pharmacology , Cadherins/drug effects , Endothelin-1/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Hypertension/metabolism , Kidney Diseases/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Biphenyl Compounds/pharmacology , Bosentan , Cadherins/metabolism , Endothelin B Receptor Antagonists/pharmacology , Endothelin-1/antagonists & inhibitors , Fibrosis , Humans , Hypertension/complications , Irbesartan , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Oligopeptides/pharmacology , Piperidines/pharmacology , Rats , Receptor, Endothelin B/metabolism , Signal Transduction , Sulfonamides/pharmacology , Tetrazoles/pharmacology , rho-Associated Kinases/metabolism
2.
J Clin Endocrinol Metab ; 99(4): E674-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24285684

ABSTRACT

CONTEXT: The molecular mechanisms of primary aldosteronism, a common cause of human hypertension, are unknown, but alterations of K(+) channels can play a key role. OBJECTIVE: The objective of the study was to investigate the following: 1) the expression of the Twik-related acid-sensitive K(+) channels (TASK) in aldosterone producing adenomas (APAs); 2) the role of TASK-2 in aldosterone synthesis; and 3) the determinants of TASK-2-blunted expression in APAs. DESIGN: We analyzed the transcriptome and the microRNA profiles of 32 consecutive APAs and investigated the protein expression and localization of TASK-2 in APA and adrenocortical cell lines (H295R and HAC15) using immunoblotting and confocal microscopy. The functional effect of TASK-2 blunted activity caused by a dominant-negative mutation on steroidogenic enzymes, and aldosterone production was also assessed. TASK-2 regulation by selected microRNA was studied by a luciferase assay. RESULTS: TASK-2 was consistently less expressed at the transcript and protein levels in APAs than in the normal human adrenal cortex. H295R cell transfection with a TASK-2 dominant-negative mutant construct significantly increased the aldosterone production by 153% and the gene expression of aldosterone synthase (CYP11B2, gene expression fold change 3.1 vs control, P < .05) and the steroidogenic acute regulatory protein (gene expression fold change 1.8 vs control, P < .05). Two microRNAs, hsa-miR-23 and hsa-miR-34, were found to decrease the TASK-2 expression by binding to the 3' untranslated region of the TASK-2 gene. CONCLUSIONS: The TASK-2 channel lower expression represents a hallmark of APA and is associated with a higher expression of hsa-miR-23 and hsa-miR-34. The ensuing blunted TASK-2 activity increased the production of aldosterone in vitro and the expression of steroidogenic acute regulatory protein and CYP11B2. Hence, the lower expression of TASK-2 channel in APA cells can explain high aldosterone secretion in human primary aldosteronism despite the suppression of angiotensin II, hypertension, and hypokalemia.


Subject(s)
Adenoma/metabolism , Adrenal Cortex Neoplasms/metabolism , Aldosterone/metabolism , Hyperaldosteronism/genetics , Potassium Channels, Tandem Pore Domain/genetics , Adenoma/genetics , Adrenal Cortex Neoplasms/genetics , Cells, Cultured , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hyperaldosteronism/metabolism , Microarray Analysis , Paraneoplastic Endocrine Syndromes/genetics , Paraneoplastic Endocrine Syndromes/metabolism
3.
PLoS One ; 6(4): e19081, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21541333

ABSTRACT

Alström Syndrome (ALMS) is a rare genetic disorder (483 living cases), characterized by many clinical manifestations, including blindness, obesity, type 2 diabetes and cardiomyopathy. ALMS is caused by mutations in the ALMS1 gene, encoding for a large protein with implicated roles in ciliary function, cellular quiescence and intracellular transport. Patients with ALMS have extensive fibrosis in nearly all tissues resulting in a progressive organ failure which is often the ultimate cause of death. To focus on the role of ALMS1 mutations in the generation and maintenance of this pathological fibrosis, we performed gene expression analysis, ultrastructural characterization and functional assays in 4 dermal fibroblast cultures from ALMS patients. Using a genome-wide gene expression analysis we found alterations in genes belonging to specific categories (cell cycle, extracellular matrix (ECM) and fibrosis, cellular architecture/motility and apoptosis). ALMS fibroblasts display cytoskeleton abnormalities and migration impairment, up-regulate the expression and production of collagens and despite the increase in the cell cycle length are more resistant to apoptosis. Therefore ALMS1-deficient fibroblasts showed a constitutively activated myofibroblast phenotype even if they do not derive from a fibrotic lesion. Our results support a genetic basis for the fibrosis observed in ALMS and show that both an excessive ECM production and a failure to eliminate myofibroblasts are key mechanisms. Furthermore, our findings suggest new roles for ALMS1 in both intra- and extra-cellular events which are essential not only for the normal cellular function but also for cell-cell and ECM-cell interactions.


Subject(s)
Apoptosis , Cell Cycle , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Proteins/metabolism , Adult , Alstrom Syndrome/genetics , Alstrom Syndrome/pathology , Apoptosis/genetics , Cell Cycle/genetics , Cell Cycle Proteins , Cell Shape , Cell Size , Female , Fibroblasts/ultrastructure , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Models, Biological , Oligonucleotide Array Sequence Analysis , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
4.
Biochim Biophys Acta ; 1793(2): 368-77, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19046997

ABSTRACT

HIPK2 has been implicated in restraining tumor progression by more than one mechanism, involving both its catalytic and transcriptional co-repressor functions. Starting from the finding that HIPK2 knockdown by RNA-interference (HIPK2i) induced significant up-regulation of HIF-1alpha mRNA and of its target VEGF in tumor cells, we evaluated the role of HIPK2 in transcriptional regulation of HIF-1alpha. We found that HIPK2 overexpression downmodulated both HIF-1alpha reporter activity and mRNA levels and showed that HIPK2 was bound in vivo to the HIF-1alpha promoter likely in a multiprotein co-repressor complex with histone deacetylase 1 (HDAC1). Thus, the HIF-1alpha promoter was strongly acetylated following HIPK2 knockdown. The HIF-1alpha-dependent VEGF transcription was evaluated by co-transfection of a dominant negative (DN) construct of HIF-1alpha that inhibited VEGF reporter activity induced by HIPK2 knockdown. HIF-1alpha and VEGF up-regulation in HIPK2i cells correlated with increased vascularity of tumor xenografts in vivo and tube formation in HUVEC in vitro. These findings provide the first evidence of HIPK2-mediated transcriptional regulation of HIF-1alpha that might play a critical role in VEGF expression.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Animals , Cell Line, Tumor , Cell Proliferation , Chromatin Assembly and Disassembly/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Repressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics
5.
J Clin Endocrinol Metab ; 94(2): 684-90, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19001524

ABSTRACT

CONTEXT: The involvement of urotensin II, a vasoactive peptide acting via the G protein-coupled urotensin II receptor, in arterial hypertension remains contentious. OBJECTIVE: We investigated the expression of urotensin II and urotensin II receptor in adrenocortical and adrenomedullary tumors and the functional effects of urotensin II receptor activation. DESIGN: The expression of urotensin II and urotensin II receptor was measured by real time RT-PCR in aldosterone-producing adenoma (n = 22) and pheochromocytoma (n = 10), using histologically normal adrenocortical (n = 6) and normal adrenomedullary (n = 5) tissue as control. Urotensin II peptide and urotensin II receptor protein were investigated with immunohistochemistry and immunoblotting. To identify urotensin II-related and urotensin II receptor-related pathways, a whole transcriptome analysis was used. The adrenocortical effects of urotensin II receptor activation were also assessed by urotensin II infusion with/without the urotensin II receptor antagonist palosuran in rats. RESULTS: Urotensin II was more expressed in pheochromocytoma than in aldosterone-producing adenoma tissue; the opposite was seen for the urotensin II receptor expression. Urotensin II receptor activation in vivo in rats enhanced (by 182 +/- 9%; P < 0.007) the adrenocortical expression of immunoreactive aldosterone synthase. CONCLUSIONS: Urotensin II is a putative mediator of the effects of the adrenal medulla and pheochromocytoma on the adrenocortical zona glomerulosa. This pathophysiological link might account for the reported causal relationship between pheochromocytoma and primary aldosteronism.


Subject(s)
Adrenal Cortex/metabolism , Adrenal Medulla/metabolism , Hyperaldosteronism/etiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Urotensins/genetics , Urotensins/physiology , Adenoma/complications , Adenoma/genetics , Adenoma/metabolism , Adrenal Cortex/drug effects , Adrenal Gland Neoplasms/complications , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adult , Animals , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyperaldosteronism/genetics , Hyperaldosteronism/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pheochromocytoma/complications , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Urotensins/metabolism , Urotensins/pharmacology
6.
Environ Toxicol Pharmacol ; 28(3): 425-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-21784038

ABSTRACT

We investigated whether chronic zidovudine (AZT) administration in rats could impair cardiac function by affecting intercellular junctions and whether vitamin C could prevent these possible effects. Rats were treated for 8 months with AZT, vitamin C, and AZT plus vitamin C. Cardiac fractional shortening (FS) was assessed by echocardiographic examination, intercellular junctions morphology was detected by electron microscopy (EM) and immunocytochemistry (ICC). AZT-treated rats showed a reduced FS that was not prevented by vitamin C. EM revealed that AZT treatment did not affect coronary endothelial intercellular junctions whereas it caused an enlargement of fascia adherens of the intercalated discs that was prevented by vitamin C. AZT treatment did not induce either alterations of gap junctions morphology or distribution of connexin-43, the major protein expressed in the gap junctions. We conclude that AZT treatment may be potentially deleterious to the heart by inducing a ROS-mediated damage of cardiac intercalated discs.

7.
J Hypertens ; 26(10): 2022-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806627

ABSTRACT

OBJECTIVE: Fibrosis is a hallmark of renal damage in several diseases, including arterial hypertension. We, therefore, investigated the role of angiotensin II, endothelin-1 and of L-type calcium channels in the development of the glomerular, vascular, and tubulointerstitial fibrosis in a model of severe angiotensin II-dependent hypertension. METHODS: Five-week-old Ren-2 transgenic rats (TGRen2) received for 4 weeks a placebo, bosentan (100 mg/kg body weight), irbesartan (50 mg/kg body weight), the ETA-selective endothelin receptor antagonist BMS-182874 (BMS; 52 mg/kg body weight), the combination of irbesartan (50 mg/kg body weight) plus BMS (52 mg/kg body weight), and nifedipine (30 mg/kg body weight). RESULTS: Glomerular volume, tubulointerstitial fibrosis, glomerular, and perivascular fibrosis were accurately quantified by histomorphometry in four-to-six sections per kidney. Glomerular fibrosis was lowered by BMS (P < 0.001), whereas tubulointerstitial fibrosis was blunted by bosentan (P < 0.001) and irbesartan (P < 0.005). Perivascular fibrosis was reduced by nifedipine and BMS. As only irbesartan and irbesartan plus BMS decreased blood pressure (P < 0.001 vs. placebo), these effects on fibrosis were independent of blood pressure. CONCLUSION: Angiotensin II and L-type calcium channels modulate fibrosis selectively in the tubulointerstitial and in the perivascular compartments, respectively. The prevention of fibrosis with ET-1 receptor antagonism in all three compartments supports a major role of ET-1 in the development of renal fibrosis.


Subject(s)
Angiotensin II/physiology , Calcium Channels, L-Type/physiology , Endothelin-1/physiology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Animals , Animals, Genetically Modified , Disease Models, Animal , Fibrosis , Kidney Failure, Chronic/pathology , Male , Rats
8.
Int J Mol Med ; 20(6): 859-64, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982695

ABSTRACT

Previous studies showed that galanin receptors are expressed in the rat adrenal, and galanin modulates glucocorticoid secretion in this species. Hence, we investigated the expression of the various galanin receptor subtypes (GAL-R1, GAL-R2 and GAL-R3) in the human adrenocortical cells, and the possible involvement of galanin in the control of cortisol secretion. Reverse transcription-polymerase chain reaction detected the expression of GAL-R1 (but not GAL-R2 and GAL-R3) in the inner zones of the human adrenal cortex. The galanin concentration dependently enhanced basal, but not ACTH-stimulated secretion of cortisol from dispersed inner adrenocortical cells (maximal effective concentration, 10(-8) M). The cortisol response to 10(-8) M galanin was abrogated by GAL-R1 immunoneutralization, and unaffected by GAL-R2 or GAL-R3 immunoneutralization. Galanin (10(-8) M) and ACTH (10(-9) M) enhanced cyclic-AMP production from dispersed cells, and the response was suppressed by the adenylate cyclase inhibitor SQ-22536 (10(-4) M). Galanin did not affect inositol triphosphate release, which, in contrast, was raised by angiotensin-II (10(-8) M). SQ-22536 and the protein kinase (PK)A inhibitor H-89 (10(-5) M) abolished the cortisol response to 10(-8) M galanin, while the phospholipase C inhibitor U-73122 and the PKC inhibitor calphostin-C were ineffective. Preincubation with pertussis toxin (Ptx) (0.5 microg/ml) partially inhibited the cortisol response to galanin. We conclude that galanin stimulates cortisol secretion from human inner adrenocortical cells, acting through GAL-R1 coupled to the adenylate cyclase/PKA-dependent signaling cascade via a Ptx-sensitive Galpha protein.


Subject(s)
Adenylyl Cyclases/metabolism , Adrenal Cortex , Galanin/metabolism , Hydrocortisone/metabolism , Receptor, Galanin, Type 1/metabolism , Second Messenger Systems/physiology , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Adult , Aged , Animals , Cyclic AMP/metabolism , Female , Humans , Male , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Receptor, Galanin, Type 1/genetics
9.
Hypertension ; 50(6): 1106-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17938379

ABSTRACT

Aldosterone-producing adenomas (APAs) are a common cause of arterial hypertension, but the underlying molecular mechanisms are unknown, although a transcriptional modulation of aldosterone synthase (CYP11B2) has been suggested. Aldosterone synthesis involves 2 main rate-limiting steps: cholesterol transport into mitochondria and CYP11B2 gene transcription. Evidence supports a role of Ca(2+)/calmodulin-dependent protein kinases (CAMKs) in the regulation of angiotensin II- and potassium-stimulated aldosterone production. CAMK-I mediates CYP11B2 transcription via cAMP response element binding protein and activating transcription factor 1 transcription factors and nuclear receptor Nur-related factor 1. CAMK-II affects cholesterol transport into mitochondria by acting on steroidogenic acute regulatory protein and/or cytoskeleton proteins. We analyzed the whole transcriptome of APAs as compared with a pool of normal human adrenocortical tissues. Based on steroidogenic enzyme gene expression profiles, we identified 2 APA subgroups: 1 featuring overexpression of CYP11B2, CAMK-I, 11-beta-hydroxylase, 3-beta-hydroxysteroid dehydrogenase, and 21-hydroxylase and the underexpression of CAMK-IIB and the other one with an opposite profile. The low CYP11B2 group exhibited a longer known duration of hypertension and a lower rate of long-term cure. Thus, aldosterone overproduction in APAs involves complex alterations of aldosterone synthesis regulation rather than simply increased aldosterone synthase gene expression. Whether the molecular signature of APA carries prognostic information is worth further investigation.


Subject(s)
Adenoma/metabolism , Adrenal Cortex Neoplasms/metabolism , Aldosterone/biosynthesis , Gene Expression Profiling , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cluster Analysis , Cytochrome P-450 CYP11B2/genetics , Humans , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , Steroid 11-beta-Hydroxylase/genetics
10.
Int Rev Cytol ; 263: 63-102, 2007.
Article in English | MEDLINE | ID: mdl-17725965

ABSTRACT

Leptin, the product of the obesity gene (ob) predominantly secreted from adipocytes, plays a major role in the negative control of feeding and acts via a specific receptor (Ob-R), six isoforms of which are known at present. Evidence has been accumulated that leptin, like other peptides involved in the central regulation of food intake, controls the function of the hypothalamic-pituitary-adrenal (HPA) axis, acting on both its central and peripheral branches. Leptin, along with Ob-R, is expressed in the hypothalamus and pituitary gland, where it modulates corticotropin-releasing hormone and ACTH secretion, probably acting in an autocrine-paracrine manner. Only Ob-R is expressed in the adrenal gland, thereby making it likely that leptin affects it by acting as a circulating hormone. Although in vitro and in vivo findings could suggest a glucocorticoid secretagogue action in the rat, the bulk of evidence indicates that leptin inhibits steroid-hormone secretion from the adrenal cortex. In keeping with this, leptin was found to dampen the HPA axis response to many kinds of stress. In contrast, leptin enhances catecolamine release from the adrenal medulla. This observation suggests that leptin activates the sympathoadrenal axis and does not appear to agree with its above-mentioned antistress action. Leptin and/or Ob-R are also expressed in pituitary and adrenal tumors, but little is known about the role of this cytokine in the pathophysiology.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Leptin/physiology , Pituitary-Adrenal System/metabolism , Animals , Humans , Receptors, Cell Surface/metabolism , Receptors, Leptin
11.
Am J Obstet Gynecol ; 196(5): 470.e1-7, 2007 May.
Article in English | MEDLINE | ID: mdl-17466707

ABSTRACT

OBJECTIVE: In vitro and in vivo studies have linked mast cell (MC) degranulation and activation with angiogenesis and neovascularization. This assumption is partially supported by the close anatomical association between MC and the vasculature and the recruitment of these cells during tumor growth. The aim of this study was to correlate the extent of angiogenesis with the number of MC expressing tryptase and leptin in human leiomyomas. STUDY DESIGN: Tissues from human leiomyomas and control specimens were investigated immunohistochemically, using murine monoclonal antibodies against the endothelial cell marker CD31, leptin, and the MC marker tryptase. RESULTS: Angiogenesis, measured as microvessel counts, was highly correlated with MC tryptase- and leptin-positive cell counts. CONCLUSION: These data suggest that angiogenesis in leiomyomas is correlated to expression of tryptase in MC granules and provide for the first time evidence of a putative role of leptin, also contained in MC secretory granules, in MC-dependent angiogenesis.


Subject(s)
Leiomyoma/physiopathology , Leptin/metabolism , Mast Cells/physiology , Neovascularization, Pathologic/physiopathology , Tryptases/metabolism , Uterine Neoplasms/physiopathology , Cell Count , Cell Degranulation , Female , Humans
12.
Int J Mol Med ; 18(6): 1097-100, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17089013

ABSTRACT

We have recently demonstrated the expression of leptin and leptin receptor (Ob-R) isoforms a, b, c, e and f in the rat seminal vesicles and prostate. The aim of the present study was to provide a semiquantitative real-time PCR estimation of leptin/Ob-R isoform mRNA expression in the seminal vesicles and individual components of rat prostate, and to ascertain the in vitro effects of leptin on prostate acid phosphatase release. The highest expression of the leptin and Ob-R genes was in the seminal vesicles and lateral prostate lobe, respectively. Of the various isoforms, Ob-Rb displayed the highest and Ob-Re the lowest expression. Leptin (10(-8) and 10(-6) M) enhanced acid phosphate release from seminal vesicles, and (10(-6) M) decreased it from the coagulating lobe. Taken together, our findings support the contention that leptin may be involved in the autocrine-paracrine functional regulation of rat seminal vesicles and prostate. The physiological relevance of the marked heterogeneity of the different prostate lobes in both their leptin/Ob-R expression and functional response to leptin remains to be addressed.


Subject(s)
Gene Expression/drug effects , Leptin/genetics , Leptin/pharmacology , Prostate/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptors, Cell Surface/genetics , Acid Phosphatase , Animals , Male , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/analysis , Rats , Receptors, Cell Surface/metabolism , Receptors, Leptin , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Int J Mol Med ; 18(6): 1101-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17089014

ABSTRACT

Neuropeptides B and W (NPB and NPW) have been identified as endogenous ligands of the G protein-coupled receptors (GPR) 7 and 8, which in humans are expressed in the hypothalamus and probably involved in the regulation of energy homeostasis and feeding behavior. GPR8 is absent in the rat, where the GPR8-like receptor (GPR8-LR) has been described. Reverse transcription-polymerase chain reaction detected the expression of NPB, NPW, GPR7 and GPR8-LR mRNAs in the hypothalamus, anterior pituitary, thyroid and parathyroid glands, pancreatic islets, adrenal glands, ovary and testis of the rat. Immunocytochemistry demonstrated the presence of NPB and NPW immunoreactivities in these same glands. Radioimmune assay showed that the bolus intraperitoneal injection of 2 nmol/100 g NPB or NPW raised the plasma levels of parathyroid hormone, corticosterone and testosterone. NPB also increased the blood concentration of thyroxine, and NPW that of ACTH and estradiol. Taken together, these findings allow us to suggest that NPB and NPW play a role in the autocrine-paracrine functional regulation of the endocrine system in the rat.


Subject(s)
Endocrine Glands/metabolism , Neuropeptides/metabolism , Receptors, Neuropeptide/metabolism , Animals , Endocrine Glands/cytology , Immunohistochemistry , Male , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Wistar
14.
Int J Mol Med ; 18(6): 1165-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17089022

ABSTRACT

The effects of four endocrine disruptors: resveratrol, diphenylolpropane (bisphenol-A; BSP), benzophenone-3 (BP3) and silymarin on the secretory and proliferative activity of rat adrenocortical cells were investigated in vitro. Resveratrol and BP3 acutely increased basal corticosterone secretion from freshly dispersed adrenocortical cells, and resveratrol and BSP enhanced ACTH-stimulated cells. The 24-h exposure to resveratrol and BP3 increased basal corticosterone production from cultured adrenocortical cells, while ACTH-stimulated secretion was increased only by resveratrol. BSP was ineffective, while silymarin decreased basal, but not ACTH-stimulated secretion. The proliferative activity of the cultured adrenocortical cells was unaffected by the tested disruptors. In conclusion, the in vitro direct effect of endocrine disruptors on adrenocortical steroidogenesis displays a great variability, which seems to depend not only on their chemical nature, but also on their dose and the duration of the exposure of the studied cells.


Subject(s)
Adrenal Cortex/drug effects , Endocrine Disruptors/pharmacology , Zona Fasciculata/drug effects , Zona Reticularis/drug effects , Adrenal Cortex/cytology , Adrenocorticotropic Hormone/pharmacology , Animals , Benzhydryl Compounds , Benzophenones/pharmacology , Cell Count , Cell Culture Techniques , Cell Separation/methods , Cells, Cultured , Corticosterone/metabolism , Dose-Response Relationship, Drug , Female , Phenols/pharmacology , Rats , Rats, Wistar , Resveratrol , Silymarin/pharmacology , Stilbenes/pharmacology , Time Factors , Zona Fasciculata/cytology , Zona Reticularis/cytology
15.
Int J Mol Med ; 18(4): 565-70, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964405

ABSTRACT

Evidence is available that some endocrine disruptors, acting as selective estrogen receptor modulators (SERMs), interfere with osteoblast differentiation and function. Therefore, we investigated whether 17beta-estradiol, bisphenol-A (BSP), silymarin, genistein, resveratrol, procymidone, linurone and benzophenone-3 (BP3) modulate differentiation of rat calvarial osteoblast-like (ROB) cells in primary in vitro culture. Disruptors were added at day 18 of culture and cells were harvested 48 h later. Real time-PCR revealed that estradiol and resveratrol enhanced osteocalcin mRNA expression in ROB cells, while other disruptors were ineffective. The expression of osteonectin and collagen-1alpha was not affected by any disruptor. Estradiol, resveratrol, genistein and BSP stimulated the proliferative activity of ROB cells. In contrast, procymidone and linurone inhibited the proliferative activity, and silymarin and BP3 were ineffective. The conclusion is drawn that i) only resveratrol is able, like estradiol, to stimulate the specialized functions of ROB cells, and ii) the proliferative activity of ROB cells is more sensitive to endocrine disruptors, some of which could probably act via a mechanism independent of their SERM activity.


Subject(s)
Collagen Type I/genetics , Estradiol/pharmacology , Osteoblasts/drug effects , Osteocalcin/genetics , Osteonectin/genetics , Stilbenes/pharmacology , Animals , Animals, Newborn , Benzhydryl Compounds , Benzophenones/pharmacology , Bridged Bicyclo Compounds/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/drug effects , Genistein/pharmacology , Linuron/pharmacology , Osteoblasts/cytology , Osteoblasts/metabolism , Phenols/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Resveratrol , Silymarin/pharmacology , Skull/cytology
16.
J Hypertens ; 24(7): 1419-27, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16794493

ABSTRACT

OBJECTIVE: The renoprotective action of angiotensin I-converting enzyme inhibitors (ACE-Is) is well established, but the role played by bradykinin (BK) remains unclear. We therefore investigated whether an enhanced BK effect on B2 receptor subtype mediated the antifibrotic effect of ACE-Is and whether neutral endopeptidase (NEP) inhibition, which can blunt BK degradation more effectively than ACE inhibition, provided further renoprotection in a rat model of angiotensin (Ang) II-dependent renal damage. METHODS: Five-week-old Ren-2 transgenic rats (TGRen2) received, for 8 weeks, a placebo, ramipril (5 mg/kg body weight) or the dual ACE + NEP inhibitor MDL 100,240 (MDL) (40 mg/kg body weight). After 4 weeks, the B2 receptor antagonist icatibant (0.5 mg/kg body weight) was administered on top of active treatment for 4 weeks to 50% of the TGRen2 rats. Blood pressure was measured weekly by a tail-cuff method and, after sacrifice, kidney weight, glomerular volume, density of glomerular profiles were measured; tubulo-interstitial fibrosis, glomerular and perivascular fibrosis were quantified by histomorphometry. RESULTS: The development of hypertension and tubulo-interstitial fibrosis was prevented by both ramipril and MDL (P = 0.0001 versus placebo); icatibant annulled the latter effect. Glomerular and perivascular fibrosis were unaffected by either ramipril or MDL alone; however, combined treatment with icatibant enhanced glomerular fibrosis (P = 0.0001 versus placebo). CONCLUSION: Enhanced BK effect on B2 subtype receptors is essential for the prevention of tubulo-interstitial fibrosis with ACE or dual ACE + NEP inhibition in TGRen2 rats.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Benzazepines/pharmacology , Kidney/pathology , Pyridines/pharmacology , Ramipril/pharmacology , Receptor, Bradykinin B2/physiology , Animals , Animals, Genetically Modified , Atrial Natriuretic Factor/drug effects , Creatinine/blood , Fibrosis/prevention & control , Hypertension/drug therapy , Kidney/drug effects , Male , Neprilysin/antagonists & inhibitors , Rats
17.
Int J Mol Med ; 17(5): 709-13, 2006 May.
Article in English | MEDLINE | ID: mdl-16596251

ABSTRACT

Adrenomedullin (AM), a potent vasodilatory hypotensive peptide, is expressed in the heart, where it is known to play a protective action. Light-microscopy immunocytochemistry (ICC) demonstrated the presence of AM immunoreactivity not only in the coronary-vessel wall and ventricular myocytes of the human and rat heart, but also in sparse voluminous cells located in the perivascular space. These cells displayed the same location of toluidine blue-positive mast cells, and electron microscopy ICC showed AM-immunogold staining over the granules of rat cardiac mast cells. The incubation of rat left ventricle fragments with the mast-cell histamine releaser compound 48/80 evidenced groups of AM-positive cells undergoing degranulation and caused an increase of approximately 50% in the AM concentration in the incubation medium. Collectively, our findings provide evidence that at least a subset of cardiac mast cells are able to synthesize and store AM, and upon stimulation to release it near coronary arterioles and venules.


Subject(s)
Mast Cells/chemistry , Myocardium/chemistry , Peptides/analysis , Adrenomedullin , Animals , Humans , Immunohistochemistry , Male , Mast Cells/drug effects , Mast Cells/ultrastructure , Microscopy, Electron , Myocardium/cytology , Rats , Rats, Sprague-Dawley , p-Methoxy-N-methylphenethylamine/pharmacology
18.
Int J Mol Med ; 15(4): 567-71, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15754015

ABSTRACT

We investigated by immunocytochemistry (ICC) the distribution in the rat heart of adrenomedullin (AM), a potent and long-lasting hypotensive peptide which is expressed in the cardiovascular system, where it is known to play a major regulatory role. Hearts were collected from adult male Sprague-Dawley rats, and were perfused for 20 min, according to the Langendorff technique, with endothelin-1 (ET-1) or the mast cell-degranulator compound 48/80. Hearts were frozen, and ICC was performed using standard techniques and a specific anti-rat AM1-50 antibody. We confirmed the presence of a low AM-immunoreactivity in cardiomyocytes and cardiac fibroblasts, as well as in endothelial and smooth muscle cells of coronary vessels. Moreover, we provided evidence of the presence in both atria and ventricles of sparse voluminous AM-positive cells, mainly located near coronary vessels. These cells had the same juxtavascular location of toluidine blue-positive mast cells and their number decreased upon acute exposure to the 48/80 compound. However, ICC showed that in these cells AM was always colocalized with atrial and brain natriuretic peptides. Moreover, AM-storing cells were also positive to MyHC-Apla2, indicating that they share some phenotypic features with immature smooth muscle cells. The number of AM-storing cells underwent a dramatic decrease in response to the potent vasoconstrictor ET-1, suggesting an acute release of stored vasodilatory AM aimed at counteracting coronary constriction. Taken together, our present findings support the hypothesis that these cells may represent a novel subset of endocrine cells, strategically located near blood vessels in the mammalian heart, where they can release vasoactive peptides.


Subject(s)
Atrial Natriuretic Factor/metabolism , Endocrine System/metabolism , Myocardium/metabolism , Natriuretic Peptide, Brain/metabolism , Peptides/metabolism , Adrenomedullin , Animals , Fluorescent Antibody Technique , Male , Rats , Rats, Sprague-Dawley
19.
Int J Mol Med ; 14(2): 165-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254759

ABSTRACT

Evidence indicates that ghrelin, an endogenous ligand of the growth hormone secretagogue receptor, controls the growth of several human and rat cell types cultured in vitro. Hence, we have investigated, by using both TUNEL and ELISA assays, the effects of 10(-8) M ghrelin on the basal apoptotic deletion rate of rat osteoblasts and thymocytes, rat and human adrenocortical cells, human umbilical vein endothelial cells, and human aldosteronoma cells cultured in vitro, as well as of the human adrenocortical carcinoma-derived cell lines NCI-H295 and SW-13. Both assays consistently showed that ghrelin did not affect apoptotic rate of normal rat and human cells, but significantly enhanced apoptotic deletion in aldosteronoma, NCI-H295 and SW-13 cell cultures. Due to the central role of apoptosis in the control of tumor growth, these findings, if confirmed in other tumor cell types, could suggest an antitumoral action of ghrelin.


Subject(s)
Apoptosis , Peptide Hormones/physiology , Adrenal Cortex/cytology , Animals , Cell Line, Tumor , Cells, Cultured/metabolism , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Ghrelin , Humans , In Situ Nick-End Labeling , Male , Osteoblasts/metabolism , Peptide Hormones/metabolism , Rats , Rats, Sprague-Dawley , Thymus Gland/cytology , Umbilical Veins/cytology
20.
Int J Mol Med ; 14(1): 71-3, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15202018

ABSTRACT

We investigated by immunocytochemistry (ICC) the acute effects of ischemia on the distribution in the rat heart of adrenomedullin (AM), a potent hypotensive peptide which is expressed in the cardiovascular system, where it is known to play a major regulatory and protective role. Hearts, collected from adult male Sprague-Dawley rats, were perfused with the Langendorff technique, and "global" ischemia was obtained by stopping perfusion for 20 min. Hearts were frozen, and ICC was performed using a specific anti-rat AM1-50 antibody and secondary peroxidase-conjugated antibodies. ICC demonstrated AM-immunoreactivity (IR) in cardiomyocytes and especially in the wall of coronary vessels. Quantitative densitometry showed that acute ischemia significantly decreased AM-IR in coronary arterioles, thereby suggesting that it markedly stimulates AM release. The conclusion is drawn that acute ischemia and ensuing hypoxia activate in the rat heart the release of AM, which by its coronarodilatory action may enhance heart blood flow.


Subject(s)
Myocardial Ischemia/metabolism , Myocardium/chemistry , Peptides/analysis , Peptides/immunology , Adrenomedullin , Animals , Immunochemistry , In Vitro Techniques , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...