Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(17)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37686217

ABSTRACT

Urotensin 2 (Uts2) is a biologically active peptide involved in the regulation of a variety of physiological and pathophysiological processes. In both the human and rat adrenal gland, the expressions of the Uts2 gene and its receptor (Uts2r) have been described. This paper focuses on the description of the hormonal control of the mRNA levels of urotensin II and its receptor in the adrenal gland of the rat, both in vitro and in vivo. The initial in vitro experiments were carried out on freshly isolated rat adrenocortical cells and their primary culture. The obtained results indicated a stimulating PKA-independent effect of ACTH on the Uts2 mRNA level in the tested cells, with no changes in the Uts2r transcript. Subsequent in vivo experiments showed that ACTH-induced adrenal growth was accompanied by an elevated level of the Uts2 mRNA, with unchanged expression of Uts2r. In the other types of in vivo gland growth studied, enucleation-induced adrenal regeneration and compensatory growth of the gland, the mRNA levels of the studied genes showed no significant differences. The only exception was hemiadrenalectomy, which led to a significant increase in Uts2 mRNA expression level 24 h after surgery. In 12-week-old rats of both sexes, gonadectomy led to a significant increase in the level of Uts2 mRNA in the adrenal gland, an effect that was prevented by sex hormones' replacement. No changes in Uts2r transcript levels were observed under these conditions. Thus, this study suggests that the regulation of Uts2 and Uts2r mRNA levels differs significantly in the rat adrenal gland. While Uts2 transcript levels appear to be mainly dependent on ACTH action, Uts2r mRNA levels are not under the control of this hormone.


Subject(s)
Secretagogues , Urotensins , Animals , Female , Humans , Male , Rats , Adrenal Glands , Adrenocorticotropic Hormone , RNA, Messenger/genetics , Urotensins/drug effects , Urotensins/genetics , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics
2.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298112

ABSTRACT

The homeostasis of the adrenal gland plays a decisive role in its proper functioning, both in non-stressful conditions and under the influence of various types of stress. This consists of interactions between all types of cells that make up the organ, including parenchymal and interstitial cells. The amount of available information on this subject in the rat adrenal glands under non-stressful conditions is insufficient; the aim of the research was to determine the expression of marker genes for rat adrenal cells depending on their location. The material for the study consisted of adrenal glands taken from intact adult male rats that were separated into appropriate zones. Transcriptome analysis by means of Affymetrix® Rat Gene 2.1 ST Array was used in the study, followed by real-time PCR validation. Expression analysis of interstitial cell marker genes revealed both the amount of expression of these genes and the zone in which they were expressed. The expression of marker genes for fibroblasts was particularly high in the cells of the ZG zone, while the highest expression of specific macrophage genes was observed in the adrenal medulla. The results of this study, especially with regard to interstitial cells, provide a so far undescribed model of marker gene expression of various cells, both in the cortex and medulla of the sexually mature rat adrenal gland. The interdependence between parenchymal and interstitial cells creates a specific microenvironment that is highly heterogeneous within the gland with respect to some of the interstitial cells. This phenomenon most likely depends on the interaction with the differentiated parenchymal cells of the cortex, as well as the medulla of the gland.


Subject(s)
Adrenal Medulla , Transcriptome , Rats , Male , Animals , Adrenal Glands/metabolism , Adrenal Medulla/metabolism , Gene Expression Profiling
3.
Biomedicines ; 11(4)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37189849

ABSTRACT

Zwilch kinetochore protein (ZWILCH) plays a key role in proper cell proliferation. The upregulation of the ZWILCH gene was observed in many types of cancers, but the association of ZWILCH with adrenocortical carcinoma (ACC) was not investigated so far. The main aim of the presented study was to verify if the enhanced level of the ZWILCH gene can be used as a diagnostic marker for ACC development and progression, as well as a predictor of survival time for ACC patients. The performed analyses included investigation of the ZWILCH expression profile in tumors with publicly available TCGA (The Cancer Genome Atlas) datasets and transcriptomic data from the Gene Expression Omnibus (GEO) database, as well as, in human biological samples of normal adrenal, adrenocortical carcinoma and in commercially available tissue microarrays. The findings demonstrate statistically significant higher ZWILCH gene expression in ACC tissue in comparison with normal adrenal glands. Furthermore, there is a strong correlation between ZWILCH upregulation and tumor mitotic rate and the probability of patient survival. The enhanced ZWILCH level is also connected with the activation of genes involved in cell proliferation and the inhibition of genes related to the immune system. This work contributes to a better understanding of the role of ZWILCH as an ACC biomarker and diagnostic tool.

4.
Front Endocrinol (Lausanne) ; 12: 713961, 2021.
Article in English | MEDLINE | ID: mdl-34276571

ABSTRACT

More than 35 years have passed since the identification of neuromedin U (NMU). Dozens of publications have been devoted to its physiological role in the organism, which have provided insight into its occurrence in the body, its synthesis and mechanism of action at the cellular level. Two G protein-coupled receptors (GPCRs) have been identified, with NMUR1 distributed mainly peripherally and NMUR2 predominantly centrally. Recognition of the role of NMU in the control of energy homeostasis of the body has greatly increased interest in this neuromedin. In 2005 a second, structurally related peptide, neuromedin S (NMS) was identified. The expression of NMS is more restricted, it is predominantly found in the central nervous system. In recent years, further peptides related to NMU and NMS have been identified. These are neuromedin U precursor related peptide (NURP) and neuromedin S precursor related peptide (NSRP), which also exert biological effects without acting via NMUR1, or NMUR2. This observation suggests the presence of another, as yet unrecognized receptor. Another unresolved issue within the NMU/NMS system is the differences in the effects of various NMU isoforms on diverse cell lines. It seems that development of highly specific NMUR1 and NMUR2 receptor antagonists would allow for a more detailed understanding of the mechanisms of action of NMU/NMS and related peptides in the body. They could form the basis for attempts to use such compounds in the treatment of disorders, for example, metabolic disorders, circadian rhythm, stress, etc.


Subject(s)
Circadian Rhythm , Energy Metabolism , Homeostasis , Metabolic Diseases/physiopathology , Neuropeptides/metabolism , Animals , Humans , Metabolic Diseases/metabolism
5.
Front Endocrinol (Lausanne) ; 11: 561370, 2020.
Article in English | MEDLINE | ID: mdl-33133015

ABSTRACT

Adropin is a multifunctional peptide hormone encoded by the ENHO (energy homeostasis associated) gene. It plays a role in mechanisms related to increased adiposity, insulin resistance, as well as glucose, and lipid metabolism. The low adropin levels are strongly associated with obesity independent insulin resistance. On the other hand, overexpression or exogenous administration of adropin improves glucose homeostasis. The multidirectional, adropin-related effects associated with the regulation of metabolism in humans also appear to be attributable to the effects of this peptide on the activity of various elements of the endocrine system including adrenal cortex. Therefore, the main purpose of the present study was to investigate the effect of adropin on proliferation and secretory activity in the human HAC15 adrenal carcinoma cell line. In this study, we obtained several highly interesting findings. First, GPR19, the main candidate sensitizer of adrenocortical cells to adropin, was expressed in HAC15 cells. Moreover, GPR19 expression was relatively stable and not regulated by ACTH, forskolin, or adropin itself. Our findings also suggest that adropin has the capacity to decrease expression levels of steroidogenic genes such as steroidogenic acute regulatory protein (StAR) and CYP11A1, which then led to a statistically significant inhibition in cortisol and aldosterone biosynthesis and secretion. Based on whole transcriptome study and research involving transforming growth factor (TGF)-ß type I receptor kinase inhibitor we demonstrated that attenuation of steroidogenesis caused by adropin is mediated by the TGF-ß signaling pathway likely to act through transactivation mechanism. We found that HAC15 cells treated with adropin presented significantly higher proliferation levels than untreated cells. Using specific intracellular inhibitors, we showed that adropin stimulate proliferation via ERK1/2 and AKT dependent signaling pathways. We have also demonstrated that expression of GPR19 is elevated in adrenocortical carcinoma in relation to normal adrenal glands. High level of GPR19 expression in adrenocortical carcinoma may constitute a negative prognostic factor of disease progression.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex/metabolism , Adrenocortical Carcinoma/metabolism , Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Adrenal Cortex/drug effects , Adrenal Cortex Neoplasms/drug therapy , Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/drug therapy , Adrenocortical Carcinoma/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Gene Regulatory Networks/physiology , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, Neurotransmitter/biosynthesis , Receptors, Neurotransmitter/genetics , Tumor Cells, Cultured
6.
Int J Mol Sci ; 20(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671654

ABSTRACT

Leptin, the first discovered adipokine, has been connected to various physiological and pathophysiological processes, including cancerogenesis. Increasing evidence confirms its influence on prostate cancer cells. However, studies on the effects of leptin on the proliferation and apoptosis of the androgen-sensitive LNCaP line of prostate cancer cells brought conflicting results. Therefore, we performed studies on the effects of high LEP concentration (1 × 10-6 M) on gene expression profile, change of selected signaling pathways, proliferation and apoptosis of LNCaP cells. RTCA (real-time cell analyzer) revealed inhibitory effect of LEP on cell proliferation, but lower LEP concentrations (10-8 and 10-10 M) did not affect cell division. Moreover, flow cytometry with a specific antibody for Cleaved PARP-1, an apoptosis marker, confirmed the activation of apoptosis in leptin-exposed LNCaP line of prostate cancer cells. Within 24 h LEP (10-6 M) increases expression of 297 genes and decreases expression of 119 genes. Differentially expressed genes (DEGs) were subjected to functional annotation and clusterization using the DAVID bioinformatics tools. Most ontological groups are associated with proliferation and apoptosis (seven groups), immune response (six) and extracellular matrix (two). These results were confirmed by the Gene Set Enrichment Analysis (GSEA). The leptin's effect on apoptosis stimulation was also confirmed using Pathview library. These results were also confirmed by qPCR method. The results of Western Blot analysis (exposure to LEP 10 min, 1, 2, 4 and 24 h) suggest (after 24 h) decrease of p38 MAPK, p44-42 mitogen-activated protein kinase and Bcl-2 phosphorylated at threonine 56. Moreover, exposure of LNCaP cells to LEP significantly stimulates the secretion of matrix metallopeptidase 7 (MMP7). Obtained results suggest activation of apoptotic processes in LNCaP cells cultured at high LEP concentration. At the same time, this activation is accompanied by inhibition of proliferation of the tested cells.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Leptin/pharmacology , Prostatic Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy
7.
Oncol Lett ; 17(3): 3599-3606, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30867803

ABSTRACT

Zinc finger protein 91 (ZFP91) gene has been recently acknowledged to possess oncogenic properties. To date, its expression has been examined only in a handful of human organs and cancer types. The aim of the present study was to characterize, for the first time, the ZFP91 expression pattern in a range of human tissues and cancer types. ZFP91 mRNA expression was examined using Cancer Survey cDNA sets. Utilized cDNA samples represented 15 human organs and 17 cancer types. ZFP91 mRNA expression was the highest in the testes and lymph nodes. It was downregulated in testis cancer, lymphoma and thyroid cancer, and upregulated in prostate cancer. Among the analyzed cancer types, ZFP91 expression was markedly elevated in sarcomas and melanoma. On a protein level, a large-scale reverse phase protein array was employed providing samples from 11 organ types and from cancers derived from these organs. ZFP91 protein expression was revealed to be generally stable across the tested samples and was only moderately elevated in breast, ovarian and pancreatic cancers. To the best of our knowledge, this is the first study to thoroughly analyze the ZFP91 expression pattern in human tissues and cancers. The obtained results provide the foundation for further work aiming to reveal its full biological significance.

8.
Int J Mol Sci ; 19(8)2018 Aug 03.
Article in English | MEDLINE | ID: mdl-30081524

ABSTRACT

Gonadotropin-inducible ovarian transcription factor-1 (Giot1) belongs to a family of fast-responsive genes, and gonadotropins rapidly induce its expression in steroidogenic cells of ovaries and testes of rats. Gonadal Giot1 gene expression is regulated by cyclic adenosine monophosphate (cAMP) -dependent protein kinase A pathway, with essential role of orphan nuclear receptor NR4A1 transcription factor (nuclear receptor subfamily 4, group A, member 1). A recent study reports that Giot1 is also expressed in adrenals, however, the mechanism of its regulation in adrenal gland is yet to be identified. Therefore, the aim of this study was to characterise the changes in Giot1 gene expression in male and female rat adrenals using wide range of in vivo and in vitro experimental models. Special emphasis was directed at the Giot1 gene regulation by ACTH and gonadotropin. In our study, we found that ACTH rapidly stimulates Giot1 expression both in vivo and in vitro. However, gonadotropin does not affect the adrenal Giot1 gene expression, presumably due to the low expression of gonadotropin receptor in adrenals. Both testosterone and estradiol administered in vivo had inhibitory effect on Giot1 gene expression in the adrenals of post-gonadectomized adult rats. Further, our studies revealed that the intracellular mechanism of Giot1 gene regulation in rat adrenals is similar to that of gonads. As in the case of gonads, the expression of Giot1 in adrenal gland is regulated by cAMP-dependent signaling pathway with essential role of the NR4A1 transcription factor. The results of our studies suggest that Giot1 may be involved in the regulation of rat adrenocortical steroidogenesis.


Subject(s)
Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/pharmacology , Chorionic Gonadotropin/pharmacology , Transcription Factors/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gonadotropins/pharmacology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics
9.
Int J Mol Sci ; 19(4)2018 Apr 07.
Article in English | MEDLINE | ID: mdl-29642441

ABSTRACT

Compensatory adrenal growth evoked by unilateral adrenalectomy (hemiadrenalectomy) constitutes one of the most frequently studied in vivo models of adrenocortical enlargement. This type of growth has been quite well characterized for its morphological, biochemical, and morphometric parameters. However, the molecular basis of compensatory adrenal growth is poorly understood. Therefore, the aim of this study was to investigate the rat adrenal transcriptome profile during the time of two previously described adrenocortical proliferation waves at 24 and 72 h after unilateral adrenalectomy. Surgical removal of the left adrenal or a sham operation was accomplished via the classic dorsal approach. As expected, the weight of the remaining right adrenal glands collected at 24 and 72 h after hemiadrenalectomy increased significantly. The transcriptome profile was identified by means of Affymetrix® Rat Gene 2.1 ST Array. The general profiles of differentially expressed genes were visualized as volcano plots and heatmaps. Detailed analyzes consisted of identifying significantly enriched gene ontological groups relevant to adrenal physiology, by means of DAVID and GOplot bioinformatics tools. The results of our studies showed that compensatory adrenal growth induced by unilateral adrenalectomy exerts a limited influence on the global transcriptome profile of the rat adrenal gland; nevertheless, it leads to significant changes in the expression of key genes regulating the circadian rhythm. Our results confirm also that regulation of compensatory adrenal growth is under complex and multifactorial control with a pivotal role of neural regulatory mechanisms and a supportive role of other components.


Subject(s)
Adrenal Glands/growth & development , Adrenalectomy/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Adrenal Glands/chemistry , Adrenal Glands/surgery , Animals , Circadian Rhythm , Disease Models, Animal , Gene Expression Regulation , Gene Ontology , Gene Regulatory Networks , Male , Organ Size , Rats
10.
Mol Med Rep ; 17(4): 6163-6173, 2018 04.
Article in English | MEDLINE | ID: mdl-29436637

ABSTRACT

Nicotinamide phosphoribosyltransferase (Nampt), also termed visfatin, catalyses the rate­limiting step in the nicotinamide adenine dinucleotide (NAD) salvage pathway. In addition to its intracellular function (iNampt), extracellular Nampt (eNampt) also affects numerous intracellular signalling pathways. The current study investigated the role of Nampt in the regulation of the hypothalamic­pituitary­adrenal (HPA) axis in rats. At 1 h after intraperitoneal administration of eNampt (4 µg/100 g) in adult male rats, serum adrenocorticotropic hormone(ACTH) and aldosterone levels remained unchanged, while corticosterone levels were notably elevated compared with the control group, as determined by ELISA. The results of reverse transcription­quantitative polymerase chain reaction (RT­qPCR) demonstrated that, in the hypothalami of eNampt­treated rats, the mRNA expression levels of Fos proto­oncogene, which is also termed c­Fos, were not significantly different compared with the control group; however, the mRNA expression levels of proopiomelanocortin (POMC) were markedly increased in the pituitary gland of eNampt­treated rats compared with the control group. Furthermore, in hypothalamic explants, ELISA results demonstrated that the addition of the eNampt protein exhibited no effect on corticotropin­releasing hormone (CRH) release into the incubation medium and prevented potassium ion­induced CRH release. Additionally, the eNampt­induced increase in ACTH output by pituitary gland explants was not statistically significant, compared with the control group. However, RT­qPCR indicated that exposure of pituitary gland explants to eNampt and CRH increased the levels of POMC mRNA expression; the effect of eNampt, but not CRH, was inhibited by FK866, which is a specific Nampt inhibitor. In primary rat adrenocortical cell cultures, eNampt exhibited no effect on basal aldosterone or corticosterone secretion, while increases in aldosterone and corticosterone levels in response to ACTH were retained. To assess the potential role of iNampt in the regulation of adrenal steroidogenesis, experiments involving a specific Nampt inhibitor, FK866, were performed. Exposure of cultured cells to FK866 notably lowered basal aldosterone and corticosterone output compared with the control group, and completely eliminated the response of cultured cells to ACTH. The results of the present study indicated that the injected eNampt may have increased the corticosterone serum levels by acting at the pituitary level. In addition, iNampt may exert a tonic stimulating effect on the secretion of aldosterone and corticosterone from rat adrenocortical cells, as normal iNampt levels were required to retain the response of cultured rat adrenocortical cells to ACTH. Thus, these data suggest an important physiological role of both iNampt and eNampt in the regulation of the HPA axis activity in the rat.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Pituitary-Adrenal System/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Aldosterone/blood , Aldosterone/metabolism , Animals , Cells, Cultured , Corticosterone/blood , Corticosterone/metabolism , Hypothalamo-Hypophyseal System/drug effects , Male , Pituitary-Adrenal System/drug effects , Rats
11.
Oncol Rep ; 39(1): 182-192, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115533

ABSTRACT

Results of studies on the expression of leptin and its receptors in the human prostate gland and human prostate cell lines are contradictory. Regarding this, we carefully reinvestigated this issue using human normal prostate (PrEC, PrSC, PrSMC) and prostate cancer (DU145, LNCaP, PC3) cell lines. Expression of leptin receptor isoforms was assessed by qPCR while the effects of leptin on cell proliferative activity was determined by real-time cell analyzer (RTCA). Expression of the leptin receptor variant 1 was not detected in LNCaP and PrSMC cell lines, but it was found in the remaining cell lines. In contrast, in all examined cell lines, isoforms 1-3 and 2 and 4 of the leptin receptor were found. The expression of isoforms 3 and 6 of the leptin receptor was observed in PC3, PrEC, PrSMC and PrSC cell lines, but not in LNCaP and DU145 cells. Expression of the leptin receptor isoforms 4-6 and 5 was not demonstrated in any of the tested cell lines. We also studied the effects of leptin on the expression of its receptor isoforms in all tested cell lines. At a wide range of concentrations, leptin did not change the expression of leptin receptor variant 1 in the DU145, PrEC and PC3 cell lines. In contrast, in the PrSC cell line, leptin significantly increased the expression of this gene. In all prostate cell lines tested, leptin did not alter the expression levels of variants 1-3 of the leptin receptor isoforms. Leptin did not alter the expression of isoforms 2 and 4 of the leptin receptor in the PC3 and LNCaP cell lines. In the DU145 and PrEC cell lines, leptin inhibited expression of these receptor isoforms while an opposite effect was noted in the PrSC cells. Leptin did not affect the expression level of variants 3 and 6 of its receptor in the PrEC and PC3 cell lines. However, in PrSMC cells, leptin inhibited the expression of variants 3 and 6 of its receptor, while in the PrSC cell line this cytokine significantly increased their expression levels. As assessed by RTCA, leptin stimulated the proliferative activity of DU145 cells, but inhibited this activity in LNCaP cells. At all concentrations tested, leptin did not change the proliferation rate of the PC3, PrEC and PrSMC cells. In contrast, leptin notably stimulated the proliferative activity of the PrSC (prostate stromal cell) cell line. Thus, our study demonstrated that in all tested human normal prostate and prostate cancer cell lines, transcription variants 4, 5 and 6 of the leptin receptor were not expressed. Leptin receptor transcription variants 1, 2 and 3 showed differential expression, which was present in the PC3, PrEC and PrSC cell lines. Our data also suggest that the stimulatory effects of leptin on proliferative activity of the studied cell lines require the expression of leptin receptor variant 1 in the affected cells.


Subject(s)
Gene Expression , Leptin/metabolism , Prostatic Neoplasms/genetics , Receptors, Leptin/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms/metabolism , RNA Isoforms/genetics
12.
Folia Histochem Cytobiol ; 55(4): 190-202, 2017.
Article in English | MEDLINE | ID: mdl-29261224

ABSTRACT

INTRODUCTION: Sirtuins are NAD dependent class III histone deacetylases. In adrenal cortex mitochondria are able to transform - via nicotinamide nucleotide transhydrogenase (NNT) - NAD into NADPH, which is required for steroidogenesis. These findings suggest that sirtuins expressed in mitochondria, Sirt3, Sirt4 and Sirt5, may be associated with adrenal steroidogenesis. Therefore, the purpose of this study was to characterize the expression of mitochondrial sirtuins (Sirt3-5) in individual compartments of rat adrenal cortex, their developmental regulation and to demonstrate whether their expression is dependent on adrenocorticotrophic hormone (ACTH) and Nampt (nicotinamide phosphoribosyltransferase also known as visfatin/PBEF), the rate-limiting enzyme in the regulation of mammalian NAD synthesis. MATERIAL AND METHODS: Studies were performed on rat adrenal glands or on primary culture of rat adrenocortical cells. Expression of mitochondrial sirtuins (Sirt3-5) was evaluated by Affymetrix microarray system or QPCR. The bulk of data were extracted from our earlier experiments which have been reanalyzed in regard to Sirt3-5 mRNAs expression levels and - if necessary - validated by QPCR. RESULTS: Sirt3-5 were expressed throughout the rat adrenal, with the highest expression level of Sirt5. The level of expression of all sirtuins is higher in the zona glomerulosa (ZG) and zona fasciculata/reticularis (ZF/R) than in the adrenal medulla. Sirt3 and Sirt5 expression levels were similar in adult male and female rats, while Sirt4 expression level was higher in females. As revealed by analysis of the available open database, no significant changes in Sirt3-5 expression levels in whole adrenal glands were observed up to week 104 of life of both male and female rats. Moreover, 60 min after intraperitoneal ACTH injection the expression level of Sirt3 in the en-tire gland was elevated while Sirt5 expression level lowered. On the other hand, chronic ACTH infusion (48 h) did not change expression of studied sirtuins. In cultured cells, ACTH greatly increased the expression levels of the Sirt4 and Sirt5. In cultured cells, Fk866 - a highly specific competitive inhibitor of Nampt - reduced expression level of Sirt5 only. In enucleation-induced regenerating rat adrenal, the expression levels of all studied sirtuins were significantly reduced in relation to the control group. Finally, in primary rat adrenal culture the FCS depletion elevates the Sirt3 and Sirt4 expression levels and downregulates Sirt5 expression. CONCLUSIONS: Sirt3-5 are expressed throughout the rat adrenal, with the highest expression levels in adrenal cortex. Performed experiments (ACTH stimulation, FCS depletion, regeneration) suggest that in the adrenal cortex, the mitochondrial Sirt5 is the primary mitochondrial sirtuin involved in regulating the biological activity of adrenocortical cells. Our results also suggest that normal levels of intracellular Nampt (iNampt) enzymatic activity are required to maintain normal (control) levels of Sirt5 mRNA in cultured cells.


Subject(s)
Adrenal Glands/metabolism , Gene Expression Regulation, Developmental , Mitochondria/enzymology , Sirtuins/genetics , Sirtuins/metabolism , Adrenal Glands/growth & development , Animals , Cells, Cultured , Female , Male , Microarray Analysis , Rats
13.
Article in English | MEDLINE | ID: mdl-28261157

ABSTRACT

Sex differences in adrenal cortex structure and function are well known in different species. In the rat, they are manifested as larger adrenal cortex and higher corticosterone secretion by females compared with males. These sex differences depend, among others, on functioning of the hypothalamic-pituitary-adrenal axis (HPA). In this aspect, it is widely accepted that testosterone exerts an inhibitory and estradiol stimulatory effect on the said axis. The molecular bases of these sex-related differences are poorly understood. Therefore, we performed studies aimed to demonstrate the effect of testosterone and estradiol on the expression of differentially regulated genes in rat adrenal gland. The classical method applied in the study-gonadectomy and gonadal hormone replacement-allows obtaining results suggesting a physiological role of the tested hormone (testosterone or estradiol) in the regulation of the specific genes. Adult male and female rats were either gonadectomized or sham operated. Half of orchiectomized rats were replaced with testosterone while ovariectomized ones with estradiol. Transcriptome was identified by means of Affymetrix® Rat Gene 2.1 ST Array. Differentially expressed genes were analyzed by means of DAVID web-based bioinformatic tools and confirmed by means of Gene Set Enrichment Analysis. For selected genes, validation of the results was performed using QPCR. Performed experiments have provided unexpected results. Contrary to expectations, in orchiectomized rats, testosterone replacement stimulates expression of numerous genes, mainly those associated with lipids and cholesterol metabolism. However, in ovariectomized animals, estradiol replacement inhibits the expression of genes, mainly those involved in intracellular signaling pathways. The physiological relevance of these findings awaits further research.

14.
Biomed Res Int ; 2016: 6963582, 2016.
Article in English | MEDLINE | ID: mdl-27975057

ABSTRACT

Novel molecular targets are being searched to aid in prostate cancer diagnosis and therapy. Recently, ZFP91 zinc finger protein has been found to be upregulated in prostate cancer cell lines. It is a potentially important oncogenic protein; however only limited data regarding its biological function and expression patterns are available. To date, ZFP91 has been shown to be a key factor in activation of noncanonical NF-κB signaling pathway as well as to be involved in HIF-1α signaling in cancer cells. The present study aimed to characterize ZFP91 expression in prostate cancer specimens. Furthermore, since our earlier reports showed discrepancies between ZFP91 mRNA and protein levels, we studied this interrelationship in LNCaP and PC-3 prostate cancer cell lines using siRNA mediated knockdown. QPCR analysis revealed marked upregulation of ZFP91 mRNA in the majority of prostate cancer specimens. Transfection of prostate cancer cells with ZFP91 siRNA resulted in a 10-fold decrease in mRNA levels. On a protein level, however, no inhibitory effect was observed over the time of the cell culture. We conclude that ZFP91 is overexpressed in prostate cancer and that potential accumulation of the ZFP91 protein in studied cells may be of importance in prostate cancer biology.


Subject(s)
Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Oncogenes , Prostatic Neoplasms/genetics , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Electrophoresis, Agar Gel , Gene Knockdown Techniques , Humans , Immunoblotting , Male , Neoplasm Grading , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transfection , Ubiquitin-Protein Ligases/genetics
15.
Folia Histochem Cytobiol ; 53(4): 333-41, 2015.
Article in English | MEDLINE | ID: mdl-26714447

ABSTRACT

INTRODUCTION: Studies on expression of orexins (OXs) and their receptors in human prostate gland and human prostatic cell lines are scanty and their results contradictory. Regarding this, we carefully reinvestigated this problem on human prostatic cell lines. MATERIAL AND METHODS: Expression of preproorexin (ppOX) (6 primer pairs), and orexin receptors 1 and 2 (OXR1, OXR2) (4 and 2 primer pairs, respectively) was assessed by conventional PCR and QPCR in human normal (PrEC, PrSc, PrSmC) and prostate carcinoma (Du145, LNCaP, and PC3) cell lines. We designed intron spanning primers and also we applied primers from earlier publications and commercially available ones. RESULTS: With the designed primer pairs, in all studied cell lines we failed to demonstrate expression of ppOX, OXR1 and OXR2 genes at the mRNA level, while reaction products were observed in control tissues (human placenta and adrenals). Primers applied in earlier studies did not form amplification products specific for preproorexin or orexin 1 receptor. Some commercially available primers for orexin receptor 1 produced false positive results. CONCLUSIONS: We found no evidence for the presence of preproorexin-orexin receptors system genes' mRNAs in human prostate cell lines. The reported premises for these genes' expression in prostate and prostatic cell lines may have arisen either from the presence of non-prostate cells included in the samples or from faulty PCR settings.


Subject(s)
Gene Expression , Orexin Receptors/biosynthesis , Orexins/biosynthesis , Prostatic Neoplasms/metabolism , Cell Line , Cell Line, Tumor , DNA Primers , Humans , Male , Orexin Receptors/genetics , Orexins/genetics , Polymerase Chain Reaction , Prostate/metabolism , Prostatic Neoplasms/genetics
16.
Folia Histochem Cytobiol ; 53(2): 133-44, 2015.
Article in English | MEDLINE | ID: mdl-26122962

ABSTRACT

INTRODUCTION: Adrenocortical activity in various species is sensitive to androgens and estrogens. They may affect adrenal cortex growth and functioning either via central pathways (CRH and ACTH) or directly, via specific receptors expressed in the cortex and/or by interfering with adrenocortical enzymes, among them those involved in steroidogenesis. Only limited data on expression of androgen and estrogen receptors in adrenal glands are available. Therefore the present study aimed to characterize, at the level of mRNA, expression of these receptors in specific components of adrenal cortex of intact adult male and female rats. MATERIAL AND METHODS: Studies were performed on adult male and female (estrus) Wistar rats. Total RNA was isolated from adrenal zona glomerulosa (ZG) and fasciculate/reticularis (ZF/R). Expression of genes were evaluated by means of Affymetrix® Rat Gene 1.1 ST Array Strip and QPCR. RESULTS: By means of Affymetrix® Rat Gene 1.1 ST Array we examined adrenocortical sex differences in the expression of nearly 30,000 genes. All data were analyzed in relation to the adrenals of the male rats. 32 genes were differentially expressed in ZG, and 233 genes in ZF/R. In the ZG expression levels of 24 genes were lower and 8 higher in female rats. The more distinct sex differences were observed in the ZF/R, in which expression levels of 146 genes were lower and 87 genes higher in female rats. Performed analyses did not reveal sex differences in the expression levels of both androgen (AR) and estrogen (ER) receptor genes in the adrenal cortex of male and female rats. Therefore matrix data were validated by QPCR. QPCR revealed higher expression levels of AR gene both in ZG and ZF/R of male than female rats. On the other hand, QPCR did not reveal sex-related differences in the expression levels of ERα, ERß and non-genomic GPR30 (GPER-1) receptor. Of those genes expression levels of ERα genes were the highest. In studied adrenal samples the relative expression of ERα mRNA was higher than ERß mRNA. In adrenals of adult male and female rats expression levels of estrogen-related receptors ERRα and ERRß were similar, and only in the ZF/R of female rats ERRγ expression levels were significantly higher than in males. We also analyzed expression profile of three isoforms of steroid 5α-reductase (Srd5a1, Srd5a2 and Srd5a3) and aromatase (Cyp19a1) and expression levels of all these genes were similar in ZG and ZF/R of male and female rats. CONCLUSIONS: In contrast to Affymetrix microarray data QPCR revealed higher expression levels of AR gene in adrenal glands of the male rats. In adrenals of both sexes expression levels of ERa, ERb, non-genomic GPR30 (GPER-1), ERR α and ERRß receptors were comparable. The obtained results suggest that acute steroidogenic effect of estrogens on corticosteroid secretion may be mediated by non-genomic GPR30.


Subject(s)
Adrenal Cortex/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Adrenal Cortex/growth & development , Animals , Aromatase/genetics , Aromatase/metabolism , Female , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Sex Characteristics
17.
J Endocrinol ; 226(1): 1-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869615

ABSTRACT

NUCB2/nesfatin and its proteolytically cleaved product nesfatin-1 are recently discovered anorexigenic hypothalamic neuroproteins involved in energy homeostasis. It is expressed both centrally and in peripheral tissues, and appears to have potent metabolic actions. NUCB2/nesfatin neurons are activated in response to stress. Central nesfatin-1 administration elevates circulating ACTH and corticosterone levels. Bilateral adrenalectomy increased NUCB2/nesfatin mRNA levels in rat paraventricular nuclei. To date, studies have not assessed the effects of nesfatin-1 stimulation on human adrenocortical cells. Therefore, we investigated the expression and effects of nesfatin-1 in a human adrenocortical cell model (H295R). Our findings demonstrate that NUCB2 and nesfatin-1 are expressed in human adrenal gland and human adrenocortical cells (H295R). Stimulation with nesfatin-1 inhibits the growth of H295R cells and promotes apoptosis, potentially via the involvement of Bax, BCL-XL and BCL-2 genes as well as ERK1/2, p38 and JNK1/2 signalling cascades. This has implications for understanding the role of NUCB2/nesfatin in adrenal zonal development. NUCB2/nesfatin may also be a therapeutic target for adrenal cancer. However, further studies using in vivo models are needed to clarify these concepts.


Subject(s)
Adrenal Cortex/cytology , Adrenal Cortex/physiology , Calcium-Binding Proteins/physiology , DNA-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Adrenal Cortex/drug effects , Adrenalectomy , Aldosterone/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Calcium Signaling , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/genetics , Female , Gene Expression , Genes, bcl-2 , Humans , Hydrocortisone/metabolism , MAP Kinase Signaling System , Mice , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Nucleobindins , Paraventricular Hypothalamic Nucleus/metabolism , Phosphoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
18.
Int J Mol Med ; 35(3): 702-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572386

ABSTRACT

Notable sex-related differences exist in mammalian adrenal cortex structure and function. In adult rats, the adrenal weight and the average volume of zona fasciculata cells of females are larger and secrete greater amounts of corticosterone than those of males. The molecular bases of these sex-related differences are poorly understood. In this study, to explore the molecular background of these differences, we defined zone- and sex-specific transcripts in adult male and female (estrous cycle phase) rats. Twelve-week-old rats of both genders were used and samples were taken from the zona glomerulosa (ZG) and zona fasciculata/reticularis (ZF/R) zones. Transcriptome identification was carried out using the Affymetrix(®) Rat Gene 1.1 ST Array. The microarray data were compared by fold change with significance according to moderated t-statistics. Subsequently, we performed functional annotation clustering using the Gene Ontology (GO) and Database for Annotation, Visualization and Integrated Discovery (DAVID). In the first step, we explored differentially expressed transcripts in the adrenal ZG and ZF/R. The number of differentially expressed transcripts was notably higher in the female than in the male rats (702 vs. 571). The differentially expressed genes which were significantly enriched included genes involved in steroid hormone metabolism, and their expression levels in the ZF/R of adult female rats were significantly higher compared with those in the male rats. In the female ZF/R, when compared with that of the males, prevailing numbers of genes linked to cell fraction, oxidation/reduction processes, response to nutrients and to extracellular stimuli or steroid hormone stimuli were downregulated. The microarray data for key genes involved directly in steroidogenesis were confirmed by qPCR. Thus, when compared with that of the males, in the female ZF/R, higher expression levels of genes involved directly in steroid hormone synthesis were accompanied by lower expression levels of genes regulating basal cell functions.


Subject(s)
Adrenal Cortex/metabolism , Transcriptome , Animals , Cluster Analysis , Computational Biology , Female , Gene Expression Profiling , Male , Molecular Sequence Annotation , Rats , Reproducibility of Results , Sex Factors , Steroids/biosynthesis
19.
Peptides ; 63: 22-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25451331

ABSTRACT

VSNL1 encodes the calcium-sensor protein visinin-like 1 and was identified previously as an upregulated gene in a sample set of aldosterone-producing adenomas. Recently, by means of microarray studies we demonstrated high expression of Vsnl1 gene in rat adrenal zona glomerulosa (ZG). Only scanty data are available on the role of this gene in adrenal function as well as on regulation of its expression by factors affecting adrenal cortex structure and function. Therefore we performed relevant studies aimed at clarifying some of the above issues. By Affymetrix(®) Rat Gene 1.1 ST Array Strip, QPCR and immunohistochemistry we demonstrated that expression levels of Vsnl1 in the rat adrenal ZG are notably higher than in the fasciculata/reticularis zone. In QPCR assay this difference was approximately 10 times higher. Expression of this gene in the rat adrenal gland or adrenocortical cells was acutely down regulated by ACTH, while chronic administration of corticotrophin or dexamethasone did not change Vsnl1 mRNA levels. In enucleation-induced adrenocortical regeneration expression levels of both Vsnl1 and Cyp11b2 were notably lowered and positively correlated. Despite these findings, the physiological significance of adrenal Vsnl1 remains unclear, and requires further investigation.


Subject(s)
Adrenal Glands/metabolism , Gene Expression , Neurocalcin/metabolism , Adrenocorticotropic Hormone/pharmacology , Adrenocorticotropic Hormone/physiology , Animals , Dexamethasone/pharmacology , Female , Glucocorticoids/pharmacology , Male , Neurocalcin/genetics , Rats, Wistar
20.
Int J Endocrinol ; 2014: 130359, 2014.
Article in English | MEDLINE | ID: mdl-25431590

ABSTRACT

Enucleation-induced adrenal regeneration is a highly controlled process; however, only some elements involved in this process have been recognized. Therefore, we performed studies on regenerating rat adrenals. Microarray RNA analysis and QPCR revealed that enucleation resulted in a rapid elevation of expression of genes involved in response to wounding, defense response, and in immunological processes. Factors encoded by these genes obscure possible priming effects of various cytokines on initiation of regeneration. In regenerating adrenals we identified over 100 up- or downregulated genes involved in adrenocortical cell proliferation. The changes were most significant at days 2-3 after enucleation and their number decreased during regeneration. For example, expression analysis revealed a notable upregulation of the growth arrest gene, Gadd45, only 24 hours after surgery while expression of cyclin B1 and Cdk1 genes was notably elevated between days 1-8 of regeneration. These changes were accompanied by changes in expression levels of numerous growth factors and immediate-early transcription factors genes. Despite notable differences in mechanisms of adrenal and liver regeneration, in regenerating adrenals we identified genes, the expression of which is well recognized in regenerating liver. Thus, it seems legitimate to suggest that, in the rat, the general model of liver and adrenal regeneration demonstrate some degree of similarity.

SELECTION OF CITATIONS
SEARCH DETAIL
...