Orexin-A stimulates 3β-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway

  • Authors:
    • Xiaocen Chang
    • Yuyan Zhao
    • Shujing Ju
    • Lei Guo
  • View Affiliations

  • Published online on: October 6, 2014     https://doi.org/10.3892/ijmm.2014.1959
  • Pages: 1523-1528
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Orexin-A is a regulatory peptide involved in the regulation of food intake, sleep-wakefulness, and it has various endocrine and metabolic functions. It orchestrates diverse central and peripheral processes through the stimulation of two G-protein coupled receptors, orexin receptor type 1 (OX1 receptor) and orexin receptor type 2 (OX2 receptor). In this study, human adrenocortical cells (NCI-H295R cells) were incubated with various concentrations of orexin-A (10-10 to 10-6 M) in vitro, and the mRNA and protein expression of OX1 receptor was determined in the cells. In addition, NCI-H295R cells treated with 10-6 M orexin-A were then treated with or without OX1 receptor specific antagonist (SB334867), AKT antagonist (PF-04691502), or a combination of both. Subsequently, cell proliferation, the cortisol content in the medium and the mRNA and protein expression expression of 3β-hydroxysteroid dehydrogenase (3β-HSD) were analyzed. The activity of the AKT signaling pathway was also determined in the NCI-H295R cells. We observed that the increase in the mRNA and protein expression of OX1 receptor was orexin-A concentration-dependent, with 10-6 M orexin-A exerting the most potent effect. Orexin-A enhanced cell proliferation and cortisol production, and increased the mRNA and protein expression of 3β-HSD in the NCI-H295R cells; however, these effects were partly blocked by the OX1 receptor antagonist, the AKT antagonist and the combination of both. Furthermore, orexin-A significantly increased the phosphorylation of AKT, with the levels of total AKT protein remaining unaltered. This effect was blocked in the presence of PF-04691502 (10-6 M), SB334867 (10-6 M) and the combination of both. On the whole, our data demonstrate that the effects of orexin-A on the survival and function of human adrenocortical cells are mediated through the AKT signaling pathway.

Introduction

Orexin-A and orexin-B, also known as hypocretin-1 and hypocretin-2, have been implicated in a wide range of central, as well as peripheral functions (14). Orexins were initially characterized as neuropeptides restricted to hypothalamic neurons in the brain that projected to nuclei involved in the control of food intake, sleep-wakefulness, neuroendocrine homeostasis and autonomic regulation (5). The functions of orexins are mediated by two membrane bound G-protein coupled receptors, the orexin receptor type 1 (OX1 receptor) and the orexin receptor type 2 (OX2 receptor). While both peptides bind with almost equal affinity to the OX2 receptor, the OX1 receptor seems to be selective for orexin-A (4). Further observations have indicated that orexins and their receptors are not restricted to the hypothalamus, but are also expressed in a few peripheral tissues (5), including the adrenal glands, gastrointestinal tract and the pancreas (4). To date, compelling evidence indicates an interaction of the orexin system with the hypothalamus-pituitary-adrenal (HPA) axis on a central, as well as peripheral level (6). Orexins (10−10 to 10−6 M) exert a stimulatory effect on glucocorticoid release, adrenocortical cell growth and, in some cases, mineralocorticoid release from the adrenal cortex of various species (713). This fact seems to be related to orexin-A (10−6 M) enhancing the expression of 3β-hydroxysteroid dehydrogenase (3β-HSD) (14). The enzymes of the 3β-HSD family that catalyze the conversion of Δ5-3β-hydroxysteroids into Δ4-3-ketosteroids are located in the endoplasmic reticulum and the mitochondrial membrane (1417). Thus, 3β-HSD plays a key role in glucocorticoid synthesis and is expressed in a number of tissues, including the adrenal glands, gonads and the brain (17). Wenzel et al (14) found that orexins stimulated glucocorticoid secretion from human adrenocortical NCI-H295R cells, and that this increase was accompanied by a simultaneous increase in the mRNA levels of 3β-HSD. However, little is known regarding the mechanisms involved in the effects of orexins leading to an increased level of 3β-HSD in adrenal cells.

AKT, also known as protein kinase B (PKB), is essential for cell survival and growth during development and carcinogenesis. AKT is a serine-threonine kinase that is regulated mainly following the activation of the second messenger, phosphatidylinositol 3-kinase (PI3K). Abundant evidence indicates that AKT is a key regulator of multiple cell survival mechanisms (1824). AKT was first characterized for its function in regulating cell proliferation and survival, which may be due to the direct or indirect effects of AKT on a number of cellular proteins. For example, AKT phosphorylates and activates mammalian target of rapamycin (mTOR) in response to growth factors and oncogenes (1921). As such, AKT plays key roles in cell survival (22), cell proliferation (23), cell growth (24) and apoptosis (22). To date, certain studies have indicated the involvement of orexins in the regulation of cell viability, cell proliferation and cortisol production (25,26). These effects can be mediated through multiple signaling pathways, including protein kinase A (PKA), protein kinase C (PKC) and mitogen-activated protein kinase (MAPK) cascade-dependent mechanisms (14,25,26). However, little is known of the ability of orexins to activate the PKB/AKT pathway in adrenal cells.

In the present study, human NCI-H295R cells were used as an adrenocortical cell model (27). The cells were exposed to various concentrations of orexin-A (10−10 to 10−6 M), in the presence of OX1 receptor antagonist, AKT antagonist or a combination of both, and cell proliferation assays were then performed to assess the effects of orexin-A on adrenocortical cell growth. Our results revealed that orexin-A significantly enhanced the expression of 3β-HSD and the production of cortisol, and increased the phosphorylation of AKT in the NCI-H295R cells. Our data present evidence for a functional role of orexin-A in human adrenocortical cells through the OX1 receptor-stimulated AKT signaling pathway.

Materials and methods

Reagents

Orexin-A was obtained from Sigma (St. Louis, MO, USA). RPMI-1640 medium and fetal bovine serum were purchased from Gibco (Grand Island, NY, USA). The AKT inhibitor, PF-04691502, was purchased from Selleck Chemicals LLC (Houston, TX, USA). The OX1 receptor-specific antagonist, SB334867, was obtained from Tocris Bioscience (Minneapolis, MN, USA). The cell proliferation enzyme-linked immunosorbent assay (ELISA) BrdU colorimetric kit was purchased from Roche Diagnostics (Penzberg, Germany). Total-AKT polyclonal antibody (ab8805), phospho-AKT (s473) polyclonal antibody (ab8932) and OX1 receptor antibody (ab68718) were obtained from Abcam (Cambridge, UK). The Cortisol Express EIA kit was purchased from Alpco (Paris, France). β-actin antibody (C4; sc-47778) and 3β-HSD antibody (37-2; sc-100466) were obtained from Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA.

Cell culture

Human NCI-H295R adrenocortical cells were obtained from the American Type Culture Collection (Manassas, VA, USA) and maintained in RPMI-1640 medium supplemented with 10% (wt/vol) fetal bovine serum, L-glutamine, penicillin (50 μg/ml) and streptomycin (100 μg/ml). The cells were grown in a humidified atmosphere containing 5% CO2 at 37°C. Prior to the experiments, the cells were grown in petri dishes in serum-free medium for 24 h. The following day, the cells (4×103 cells/well in 96-well plates, 5×105 cells/well in 6-well plates) were treated with various concentrations of orexin-A (0, 10−10, 10−8 and 10−6 M), or 10−6 M orexin-A with either SB334867 or PF-04691502, or a combination of both.

Cell proliferation assays

The adrenocortical NCI-H295R cells were seeded (2×103 cells/well) in 96-well plates and cultured for 24 h. To synchronize the cell cycle, the cells were serum-deprived for 24 h and then treated with the test agents for a further 24 h. BrdU incorporation into DNA was measured using the Cell Proliferation ELISA BrdU colorimetric kit (Roche Diagnostics). The cells were incubated with BrdU fresh medium at 37°C and 5% CO2 for 12 h and fixed with 200 μl of fixative/denaturing solution for 30 min at room temperature. Peroxidase-conjugated BrdUrd antibody was then added to each well followed by incubation for 1 h. After washing thoroughly, the bound peroxidase-conjugated BrdUrd antibody was quantified with peroxidase substrate tetramethylbenzidine. Finally the BrdUrd absorbance was measured at 440 nm using an ELISA plate reader (BioTek Instruments, Winooski, VT, USA). A control without cells was used to measure the background absorbance of the medium and was subtracted from the results.

Cortisol measurements

For cortisol release experiments, the NCI-H295R cells were cultured in 6-well plates until the cells were at approximately 80–85% confluence. The cells were serum starved overnight, then washed and incubated in fresh serum-free medium containing various concentrations of orexin-A and the different inhibitors for 24 h. At the end of the incubation period, the supernatant was removed and snap-frozen immediately in liquid nitrogen until the cortisol measurements were taken. Cortisol levels were assessed using the ELISA kit according to the manufacturer’s instructions.

Total RNA preparations and reverse transcription quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the NCI-H295R cells using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). The mRNA expression of OX1 receptor and OX2 receptor was detected by real-time PCR using TaqMan reagents (Takara, Otsu, Japan). The following specific primers were used: OX1 receptor forward, 5′-TGC GGC CAA CCC TAT CAT CTA-3′ and reverse, 5′-ACC GGC TCT GCA AGG ACAA-3′; OX2 receptor forward, 5′-ATC GCA GGG TAT ATC ATC GTG TTC-3′ and reverse, 5′-TGA CTG TCC TCA TGT GGT GGT TC-3′; 3β-HSD forward, 5′-AGC AAA AAG ATG GCC GAG AA-3′ and reverse, 5′-GGC ACA AGT ATG CAA TGT GCC-3′. As an internal control for reverse transcription (RT) and reaction efficiency, the amplification of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA was carried out in parallel for each sample. The following specific primers were used: GAPDH forward, 5′-GGC ACA GTC AAG GCT GAG AAT G-3′ and reverse, 5′-ATG GTG GTG AAG ACG CCA GTA-3′. The PCR reactions were carried out using the following conditions: 95°C for 30 sec, then 40 cycles of 95°C for 5 sec, 60°C for 30 sec, and 95°C for 15 sec. All primers and TaqMan probes specific to OX1 receptor, OX2 receptor, 3β-HSD and GAPDH were designed using Primer Premier 5.0 software (Premier Biosoft International, Palo Alto, CA, USA).

Protein preparations and western blot analysis

The NCI-H295R cells were washed with cold phosphate-buffered saline (PBS) and harvested in RIPA buffer containing protease inhibitors. The cell lysates were incubated on ice for 30 min and were collected and centrifuged at 12,000 × g for 10 min at 4°C. The supernatants were collected and mixed with 5X loading buffer, then denatured by boiling for 10 min. The samples were separated by sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE) and transferred onto PVDF membranes at 60 V for 2.5 h in transfer buffer containing 20 mM Tris, 150 mM glycine and 20% methanol. The membranes were incubated in non-fat dry milk for 120 min at room temperature, and then washed 3 times with TBST for 30 min, then incubated with primary antibody against OX1 receptor at a 1:250 dilution, phospho/total-AKT at a 1:1,000 dilution or antibody against 3β-HSD at a 1:1,000 dilution in TBST overnight at 4°C. The membranes were washed and incubated with a secondary antibody (source: rabbit) for 1.5 h at room temperature, and then washed 3 times with TBST for 30 min. Protein was visualized using the ECL method. Band densities were measured using Quantity One software.

Statistical analysis

The results are expressed as the means ± standard error of the mean (SEM) and differences between the means were analyzed by one-way analysis of variance (ANOVA). A value of P≤0.05 was considered to indicate a statistically significant difference.

Results

Orexin-A receptor expression in NCI-H295R cells

RT-qPCR assays demonstrated that OX1 receptor mRNA was expressed in the NCI-H295R cells (Fig. 1A). However, OX2 receptor mRNA was not detectable under the same conditions (data not shown). Orexin-A (10−10, 10−8 and 10−6 M) induced a significant increase in the mRNA and protein levels of OX1 receptor in a dose-dependent manner (Fig. 1). Treatment with orexin-A increased OX1 receptor protein expression in the NCI-H295R cells, an increase that was dependent upon the concentration of orexin-A, with 10−6 M of orexin-A exerting the most potent effect (Fig. 1B). This increase in expression was attenuated in the presence of 10−6 M SB334867, a high-affinity, OX1 receptor-specific, non-peptide antagonist (Fig. 1B).

Orexin-A enhances the proliferation of NCI-H295R cells

To determine the effects of orexin-A on cell survival, as well as the involvement of the AKT signaling pathway in the effects of orexin-A in the NCI-H295R cells, we employed BrdU incorporation assay to examine cell proliferation. The NCI-H295R cells were treated with various concentrations (0, 10−10, 10−8 and 10−6 M) of orexin-A; the promoting effects on cell proliferation were dose-dependent. Concentrations of 10−6 and 10−8 M of orexin-A led to a 0.8- and 0.6-fold increase in cell proliferation, respectively. However, these effects were partly blocked when the cells were treated with 10−6 M orexin-A and the OX1 receptor antagonist, SB334867 (10−6 M), or the AKT antagonist, PF-04691502 (10−6 M), as well as with a combination of both antagonists. Taken together, these data suggest that AKT participates in the orexin-A-induced stimulation of the proliferation of NCI-H295R cells (Fig. 2).

Orexin-A induces cortisol secretion from NCI-H295R cells

To determine whether the production of the cortisol is affected in orexin-A-stimulated NCI-H295R cells, cortisol levels in the culture medium were assessed using an ELISA kit. The dose-dependent effects of orexin-A on the cortisol content in the medium were determined from the cell culture supernatants. The effects of 10−6 and 10−8 M orexin-A reached statistical significance, increasing cortisol secretion by 1.0- and 0.5-fold, respectively compared to the controls (untreated cells). This affect was blocked in the presence of PF-04691502 (10−6 M), SB334867 (10−6 M) and the combination of both antagonists (Fig. 3).

Effects of orexin-A on 3β-HSD mRNA and protein expression

Follwoing starving overnight in serum-free medium, the NCI-H295R cells were incubated with various concentrations (0, 10−10, 10−8 and 10−6 M) of orexin-A, and the cells were then treated with 10−6 M orexin-A and the OX1 receptor antagonist, SB334867 (10−6 M), or the AKT antagonist, PF-04691502 (10−6 M), or a combination of both antagonists. Treatment with orexin-A increased the mRNA and protein expression levels of 3β-HSD in the NCI-H295R cells, an increase that was dependent on the concentration of orexin-A, with 10−6 M of orexin-A exerting the most potent effect. Concentrations of 10−6 M and 10−8 M orexin-A led to a 2.1- and 2.4-fold increase in the mRNA levels, respectively (Fig. 4A). As regards protein expression, 10−6 M orexin-A and 10−8 M orexin-A induced a 1.8- and 1.2-fold increase in expression, respectively compared to the controls (Fig. 4B). This increase was attenuated in the presence of 10−6 M SB334867 or 10−6 M PF-04691502, or the combination of both antagonists.

Orexin-A signals through the AKT pathway

To determine wether the orexin-A-stimulation of NCI-H295R cells induces the activation of AKT, the NCI-H295R cells were stimulated with various concentrations (0, 10−6, 10−8 and 10−10 M) of orexin-A. The data demonstrated a specific increase in the levels of phospo-AKT in the NCI-H295R cells treated with 10−6 and 10−8 M orexin-A, increasing by 0.7- and 0.45-fold, respectively, compared to the untreated controls (Fig. 5). The levels of total AKT, however, remained unaltered by treatment. In addition, the relative increase in AKT activation in response to orexin-A was abolished by treatment with AKT antagonist (PF-04691502, 10−6 M), OX1 receptor antagonist (SB334867, 10−6 M), as well as the combination of both antagonsits (Fig. 5). Above all, these data suggest that the regulation of the AKT signaling pathway is intimately associated with the orexin-A-stimulated NCI-H295R cell survival through the OX1 receptor.

Discussion

Studies have indicated an association between the hypocretin/orexin system and the HPA axis on a central, as well as peripheral level (7). In this study, we dertermined the effects of orexin-A on the expression of OX1 receptor and the proliferation of human adrenocortical cells. Consistent with the study by Blanco et al (28), we did not observe the expression of OX2 receptor in the NCI-H295R cells. Furthermore, we found a marked increase in cortisol production and 3β-HSD expression following the stimulation of the NCI-H295R cells with orexin-A, that was associated with an increased activity of AKT. This finding indicates that another signaling pathway, the AKT pathway, partly regulates the survival and functions of adrenocortical cells stimulated with orexin-A through the OX1 receptor.

Previous studies addressing the effects of orexins on adrenal function focused on the effects of the HPA axis. One of the first in vivo studies described an increase in both adrenocorticotropic hormone (ACTH) and corticosterone plasma levels 1–2 h following a single systemic administration of orexin-A in rats, while orexin-B had no stimulating effect (9). Other studies clearly demonstrated an additional direct action of orexins on the adrenal cortex without the involvement of the HPA axis (7,12). Orexin-A enhances the production of mineralocorticoids, glucocorticoids and androgens in the adrenal glands (29). Furthermore, studies have demonstrated the important role of orexin-A in adrenocortical cell proliferation (8,12). However, data on whether orexins regulate the growth of carcinoma cells are inconlcusive. Certain studies have found that orexins promote the growth of cancer cells (8,10), athough others have shown inhibitory effects (30,31). The different cell types used may be the reason for these conflicting results. Consistent with previous studies, we found that orexin-A stimulated NCI-H295R cell proliferation (8,10) and promoted the release of cortisol in a dose-dependent manner, while the effects were blunted by co-treatment with the OX1 receptor antagonist, SB334867. The synthesis of glucocorticoids in human adrenal glands is achieved by the selective expression of some steroid-synthesizing enzymes, such as 3β-HSD. As previously reported, the selective upregulation of 3β-HSD mRNA expression following prolonged stimulation with orexins indicated the existence of a specific pathway for the transcriptional regulation of orexins (14). In this study, we found the highest cortisol synthesis rate after 24 h of treatment with orexin-A, possibly due to the stimulation of the expression of 3β-HSD and the proliferation of adrenocortical cells. This was consistent with the results of a previous study by Wenzel et al, who demonstrated that orexins increased the expression of steroidogenic enzymes at the transcriptional level and that orexins played a role in the long-term regulation of adrenal steroid production (14).

As is well known, among mechanisms controlling the cell growth and survival, the PI3K signaling pathway is often activated. The serine/threonine kinase AKT, acting downstream of PI3K signaling, is a key regulator of multiple survival routes (3237). Previous studies focused on the effects of orexin-A on the MAPK pathway (6,25). Göncz et al (38) found that orexin-A modulated glucagon secretion and gene expression through the PI3K/AKT-dependent pathway in clonal pancreatic A-cells (InR1-G9 cells). They found an increase in the phosphorylation of AKT and phosphoinositide-dependent kinase-1 (PDK-1) proteins in response to treatment with orexin-A (10−5 M) in InR1-G9 cells. Our study demonstrated that orexin-A (10−10 to 10−6 M) stimulated NCI-H295R cell proliferation and promoted the release of cortisol and increased the expression of 3β-HSD. In addition we observed a specific increase in the levels of phospho-AKT in the NCI-H295R cells treated with 10−6 and 10−8 M orexin-A. The levels of total AKT, however, remained unaltered by treatment. The increase in AKT phosphorylation may be involved in the regulation of the expression of 3β-HSD. However, these effects were partly blocked by co-treatment with the OX1 receptor antagonist, SB334867, or the AKT antagonist, PF-04691502, as well as with the combination of both antagonists. With respect to the concentration of orexin-A, it is possible that the cell type may be an important factor contributing to the physiological effects of orexin-A. Further studies are required in order to further determine to what extent the cell type plays a role in the effects of orexin-A. Taken together, these data suggest that orexin-A stimulates 3β-HSD expression and cortisol production in human adrenocortical cells through the OX1 receptor mediated through the AKT pathway.

In conclusion, we demonstrate that the AKT signaling pathway is involved in the orexin-A stimulated increase in the synthesis of cortisol in human adrenocortical cells. Although this interaction of orexins with adrenocortical cell functions, particularly with glucocorticoid production, has now bee established, more comprehensive and specific mechanisms remain to be elucidated to clarify the nature of the signaling pathway. On the whole, orexins, together with leptin, may comprise a counter-regulatory system that controls body weight and energy homeostasis through the regulation of adrenocortical steroid production.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (grant nos. 30872724, 81071460 and 81271996). We are thankful to the China Medical University Affiliated Hospital Laboratory Center for kindly providing the necessary equipment.

Abbreviations:

OX1 receptor

orexin receptor type 1

OX2 receptor

orexin receptor type 2

AKT/PKB

protein kinase B

MAPK

mitogen-activated protein kinase

PI3K

phosphatidylinositol 3-kinase

PKA

protein kinase A

PKC

protein kinase C

3β-HSD

3β-hydroxysteroid dehydrogenase

References

1 

de Lecea L, Kilduff TS, Peyron C, Gao X, Foye PE, Danielson PE, Fukuhara C, Battenberg EL, Gautvik VT, Bartlett FS II, Frankel WN, van den Pol AN, Bloom FE, Gautvik KM and Sutcliffe JG: The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci USA. 95:322–327. 1998.PubMed/NCBI

2 

Heinonen MV, Purhonen AK, Mäkelä KA and Herzig KH: Functions of orexins in peripheral tissues. Acta Physiol (Oxf). 192:471–485. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Matsuki T and Sakurai T: Orexins and orexin receptors: from molecules to integrative physiology. Results Probl Cell Differ. 46:27–55. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Sakurai T1, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richardson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ and Yanagisawa M: Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled eceptors that regulate feeding behavior. Cell. 92:573–585. 1998. View Article : Google Scholar

5 

Voisin T, Rouet-Benzineb P, Reuter N and Laburthe M: Orexins and their receptors: structural aspects and role in peripheral tissues. Cell Mol Life Sci. 60:72–87. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Kagerer SM and Jöhren O: Interactions of orexins/hypocretins with adrenocortical functions. Acta Physiol (Oxf). 198:361–371. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Malendowicz LK, Hochol A, Ziolkowska A, Nowak M, Gottardo L and Nussdorfer GG: Prolonged orexin administration stimulates steroid-hormone secretion, acting directly on the rat adrenal gland. Int J Mol Med. 7:401–404. 2001.PubMed/NCBI

8 

Malendowicz LK, Jedrzejczak N, Belloni AS, Trejter M, Hochol A and Nussdorfer GG: Effects of orexins A and B on the secretory and proliferative activity of immature and regenerating rat adrenal glands. Histol Histopathol. 16:713–717. 2001.PubMed/NCBI

9 

Malendowicz LK, Tortorella C and Nussdorfer GG: Orexins stimulate corticosterone secretion of rat adrenocortical cells, through the activation of the adenylate cyclase-dependent signaling cascade. J Steroid Biochem Mol Biol. 70:185–188. 1999. View Article : Google Scholar

10 

Mazzocchi G, Malendowicz LK, Gottardo L, Aragona F and Nussdorfer GG: Orexin A stimulates cortisol secretion from human adrenocortical cells through activation of the adenylate cyclase-dependent signaling cascade. J Clin Endocrinol Metab. 86:778–782. 2001. View Article : Google Scholar

11 

Nanmoku T, Isobe K, Sakurai T, Yamanaka A, Takekoshi K, Kawakami Y, Goto K and Nakai T: Effects of orexin on cultured porcine adrenal medullary and cortex cells. Regul Pept. 104:125–130. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Spinazzi R, Rucinski M, Neri G, Malendowicz LK and Nussdorfer GG: Preproorexin and orexin receptors are expressed in cortisol-secreting adrenocortical adenomas, and orexins stimulate in vitro cortisol secretion and growth of tumor cells. J Clin Endocrinol Metab. 90:3544–3549. 2005. View Article : Google Scholar

13 

Ziolkowska A, Spinazzi R, Albertin G, Nowak M, Malendowicz LK, Tortorella C and Nussdorfer GG: Orexins stimulate glucocorticoid secretion from cultured rat and human adrenocortical cells, exclusively acting via the OX1 receptor. J Steroid Biochem Mol Biol. 96:423–429. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Wenzel J, Grabinski N, Knopp CA, Dendorfer A, Ramanjaneya M, Randeva HS, Ehrhart-Bornstein M, Dominiak P and Jöhren O: Hypocretin/orexin increases the expression of steroidogenic enzymes in human adrenocortical NCI H295R cells. Am J Physiol Regul Integr Comp Physiol. 297:R1601–R1609. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Hayes HM Jr and Wilson GP: Hormone-dependent neoplasms of the canine perianal gland. Cancer Res. 37:2068–2071. 1977.PubMed/NCBI

16 

Sauer LA, Chapman JC and Dauchy RT: Topology of 3 beta-hydroxy-5-ene-steroid dehydrogenase/delta 5-delta 4-isomerase in adrenal cortex mitochondria and microsomes. Endocrinology. 134:751–759. 1994.PubMed/NCBI

17 

Zhao HF, Labrie C, Simard J, de Launoit Y, Trudel C, Martel C, Rhéaume E, Dupont E, Luu-The V, Pelletier G and Labriel F: Characterization of rat 3 beta-hydroxysteroid dehydrogenase/delta 5- delta 4 isomerase c DNAs and differential tissue specific expression of the corresponding mRNAs in steroidogenic and peripherial tissues. J Biol Chem. 266:583–593. 1991.

18 

van Blitterswijk WJ and Verheij M: Anticancer mechanisms and clinical application of alkylphospholipids. Biochim Biophys Acta. 1831:663–674. 2013.PubMed/NCBI

19 

Fu L, Kim YA, Wang X, Wu X, Yue P, Lonial S, Khuri FR and Sun SY: Perifosine inhibits mammalian target of rapamycin signaling through facilitating degradation of major components in the mTOR axis and induces autophagy. Cancer Res. 69:8967–8976. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Hay N and Sonenberg N: Upstream and downstream of mTOR. Genes Dev. 18:1926–1945. 2004. View Article : Google Scholar

21 

Richardson CJ, Schalm SS and Blenis J: PI3-kinase and TOR: PIKTORing cell growth. Semin Cell Dev Biol. 15:147–159. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Hsieh AC, Truitt ML and Ruggero D: Oncogenic AKTivation of translation as a therapeutic target. Br J Cancer. 105:329–336. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Sale EM and Sale GJ: Protein kinase B: signalling roles and therapeutic targeting. Cell Mol Life Sci. 65:113–127. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Blandino-Rosano M, Chen AY, Scheys JO, Alejandro EU, Gould AP, Taranukha T, Elghazi L, Cras-Méneur C and Bernal-Mizrachi E: mTORC1 signaling and regulation of pancreatic β-cell mass. Cell Cycle. 11:1892–1902. 2012.

25 

Ramanjaneya M, Conner AC, Chen J, Kumar P, Brown JE, Jöhren O, Lehnert H, Stanfield PR and Randeva HS: Orexin-stimulated MAP kinase cascades are activated through multiple G-protein signalling pathways in human H295R adrenocortical cells: diverse roles for orexins A and B. J Endocrinol. 202:249–261. 2009. View Article : Google Scholar

26 

Ramanjaneya M, Conner AC, Chen J, Stanfield PR and Randeva HS: Orexins stimulate steroidogenic acute regulatory protein expression through multiple signaling pathways in human adrenal H295R cells. Endocrinology. 149:4106–4115. 2008. View Article : Google Scholar

27 

Rainey WE, Saner K and Schimmer BP: Adrenocortical cell lines. Mol Cell Endocrinol. 228:23–38. 2004. View Article : Google Scholar

28 

Blanco M, García-Caballero T, Fraga M, Gallego R, Cuevas J, Forteza J, Beiras A and Diéguez C: Cellular localization of orexin receptors in human adrenal gland, adrenocortical adenomas and pheochromocytomas. Regul Pept. 104:161–165. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Payne AH and Hales DB: Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr Rev. 25:947–970. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Rouet Benzineb P, Rouyer Fessard C, Jarry A, Avondo V, Pouzet C, Yanagisawa M, Laboisse C, Laburthe M and Voisin T: Orexins acting at native OX(1) receptor in colon cancer and neuroblastoma cells or at recombinant OX(1) receptor suppress cell growth by inducing apoptosis. J Biol Chem. 279:45875–45886. 2004.PubMed/NCBI

31 

Biegańska K, Sokołowska P, Jöhren O and Zawilska JB: Orexin A suppresses the growth of rat C6 glioma cells via a caspase-dependent mechanism. J Mol Neurosci. 48:706–712. 2012.PubMed/NCBI

32 

Alessandro R and Kohn EC: Signal transduction targets in invasion. Clin Exp Metastasis. 19:265–273. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Sachdev P, Jiang YX, Li W, Miki T, Maruta H, Nur-E-Kamal MS and Wang LH: Differential requirement for Rho family GTPases in an oncogenic insulin-like growth factor-I receptor-induced cell transformation. J Biol Chem. 276:26461–26471. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Nguyen KT, Wang WJ, Chan JL and Wang LH: Differential requirements of the MAP kinase and PI3 kinase signaling pathways in Src- versus insulin and IGF-1 receptors-induced growth and transformation of rat intestinal epithelial cells. Oncogene. 19:5385–5397. 2000. View Article : Google Scholar

35 

Sachdev LP, Zeng and Wang LH: Distinct role of phosphatidylinositol 3-kinase and Rho family GTPases in Vav3-induced cell transformation, cell motility, and morphological changes. J Biol Chem. 277:17638–17648. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Nguyen KT, Zong CS, Uttamsingh S, Sachdev P, Bhanot M, Le MT, Chan JL and Wang LH: The role of phosphatidylinositol 3-kinase, rho family GTPases, and STAT3 in Ros-induced cell transformation. J Biol Chem. 277:11107–11115. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Arboleda MJ, Lyons JF, Kabbinavar FF, Bray MR, Snow BE, Ayala R, Danino M, Karlan BY and Slamon DJ: Overexpression of AKT2/protein kinase Bbeta leads to up-regulation of beta1 integrins, increased invasion, and metastasis of human breast and ovarian cancer cells. Cancer Res. 63:196–206. 2003.PubMed/NCBI

38 

Göncz E, Strowski MZ, Grötzinger C, Nowak KW, Kaczmarek P, Sassek M, Mergler S, El-Zayat BF, Theodoropoulou M, Stalla GK, Wiedenmann B and Plöckinger U: Orexin-A inhibits glucagon secretion and gene expression through a Foxo1-dependent pathway. Endocrinology. 149:1618–1626. 2008.PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 34 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chang X, Zhao Y, Ju S and Guo L: Orexin-A stimulates 3β-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway. Int J Mol Med 34: 1523-1528, 2014
APA
Chang, X., Zhao, Y., Ju, S., & Guo, L. (2014). Orexin-A stimulates 3β-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway. International Journal of Molecular Medicine, 34, 1523-1528. https://doi.org/10.3892/ijmm.2014.1959
MLA
Chang, X., Zhao, Y., Ju, S., Guo, L."Orexin-A stimulates 3β-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway". International Journal of Molecular Medicine 34.6 (2014): 1523-1528.
Chicago
Chang, X., Zhao, Y., Ju, S., Guo, L."Orexin-A stimulates 3β-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway". International Journal of Molecular Medicine 34, no. 6 (2014): 1523-1528. https://doi.org/10.3892/ijmm.2014.1959