Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Biomolecules ; 11(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34439753

ABSTRACT

There is a pressing need for molecular targets and biomarkers in gastric cancer (GC). We aimed at identifying aberrations in L-arginine metabolism with therapeutic and diagnostic potential. Systemic metabolites were quantified using mass spectrometry in 293 individuals and enzymes' gene expression was quantified in 29 paired tumor-normal samples using qPCR and referred to cancer pathology and molecular landscape. Patients with cancer or benign disorders had reduced systemic arginine, citrulline, and ornithine and elevated symmetric dimethylarginine and dimethylamine. Citrulline and ornithine depletion was accentuated in metastasizing cancers. Metabolite diagnostic panel had 91% accuracy in detecting cancer and 70% accuracy in differentiating cancer from benign disorders. Gastric tumors had upregulated NOS2 and downregulated ASL, PRMT2, ORNT1, and DDAH1 expression. NOS2 upregulation was less and ASL downregulation was more pronounced in metastatic cancers. Tumor ASL and PRMT2 expression was inversely related to local advancement. Enzyme up- or downregulation was greater or significant solely in cardia subtype. Metabolic reprogramming in GC includes aberrant L-arginine metabolism, reflecting GC subtype and pathology, and is manifested by altered interplay of its intermediates and enzymes. Exploiting L-arginine metabolic pathways for diagnostic and therapeutic purposes is warranted. Functional studies on ASL, PRMT2, and ORNT1 in GC are needed.


Subject(s)
Arginine/metabolism , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/metabolism , Aged , Argininosuccinate Lyase/biosynthesis , Cell Differentiation , Citrulline/metabolism , DNA, Complementary/metabolism , Female , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Mass Spectrometry , Metabolomics , Middle Aged , Mitochondrial Membrane Transport Proteins/biosynthesis , Neoplasm Metastasis , Nitric Oxide Synthase Type II , Ornithine/metabolism , Polymerase Chain Reaction , Protein-Arginine N-Methyltransferases/biosynthesis , Reproducibility of Results , Stomach Neoplasms/drug therapy , Transcriptome
2.
Biochem Biophys Res Commun ; 508(3): 973-979, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30551877

ABSTRACT

XAF1 is a tumor suppressor gene with low or absent expression in cancer. Since transcriptional reactivation or ectopic-mediated expression of XAF1 inhibits tumor growth, it is of great interest to elucidate the molecular mechanisms leading to XAF1 silencing. YY1 is a transcription factor that acts as a repressor or an activator to modulate several cancer-associated cellular processes. Both YY1 and XAF1 have key roles in prostate cancer (PCa) progression and are associated with worse clinical outcomes. To assess whether YY1 regulates the transcriptional activation of the XAF1 gene, we performed gene-reporter assays coupled with site-directed mutagenesis, which showed that YY1 is able to mediate XAF1 silencing. Concordantly, ChIP-qPCR assays showed that YY1 interacts with the XAF1 promoter in PC3 cells that lacks XAF1 expression. This association was lost after exposure to epigenetic modulators that induce XAF1 expression. Further supporting the YY1's repressive role, we found transcriptional reactivation of the XAF1 gene by YY1 downregulation. As expected by previous reports showing that HDAC1 is needed for YY1-mediated repressive actions, we observed XAF1 re-expression after either inhibition or downregulation of the HDAC1 gene. Finally, expression data retrieved from the TCGA consortium showed that PCa samples presented lower XAF1 and higher HDAC expression levels than normal tissues. Thus, our results support a model in which YY1 is able to silence tumor suppressor genes such as XAF1 through HDAC1 in PCa.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , YY1 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Binding Sites , Cell Line, Tumor , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Repressor Proteins/metabolism , YY1 Transcription Factor/genetics
3.
Tumour Biol ; 39(3): 1010428317694536, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28351329

ABSTRACT

The chemotherapy with fluorouracil is not always effective, in which some breast cancer cells may survive the fluorouracil treatment through enhanced autophagy. Crocetin is the major constituent of saffron, a Chinese traditional herb, which has recently found to have multiple pharmacological effects, including anticancer. However, the effects of Crocetin on the outcome of fluorouracil therapy for breast cancer have not been studied. Here, we showed that fluorouracil treatment inhibited the growth of breast cancer cells, in either a Cell Counting Kit-8 assay or an MTT assay. Inhibition of autophagy further suppressed breast cancer cell growth, suggesting that the breast cancer cells increased autophagic cell survival during fluorouracil treatment. However, Crocetin significantly increased the suppressive effects of fluorouracil on breast cancer cell growth, without affecting either cell apoptosis or autophagy. Inhibition of autophagy at the presence of Crocetin partially abolished the suppressive effects on breast cancer cell growth, suggesting that Crocetin may increase autophagic cell death in fluorouracil-treated breast cancer cells. Furthermore, Crocetin decreased Beclin-1 levels but increased ATG1 levels in fluorouracil-treated breast cancer cells. Together, these data suggest that Crocetin may shift autophagic cell survival to autophagic cell death in fluorouracil-treated breast cancer cells, possibly through modulation of the expression of ATG1 and Beclin-1.


Subject(s)
Breast Neoplasms/drug therapy , Carotenoids/administration & dosage , Drug Synergism , Fluorouracil/administration & dosage , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/biosynthesis , Beclin-1 , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , MCF-7 Cells , Medicine, Chinese Traditional , Vitamin A/analogs & derivatives , Xenograft Model Antitumor Assays
4.
Int J Mol Med ; 37(2): 501-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26707911

ABSTRACT

Arctium lappa fruit has been used in traditional medicine, and it is known to exert beneficial effects, such as antioxidant, anti-inflammatory and anticancer effects. However, the effects of the Arctium lappa fruit on the allergic response remain unknown. In this study, we evaluated the anti-allergic effects of Arctium lappa fruit extract (AFE) and its fermented form (F-AFE) using immunoglobulin E (IgE)-activated RBL­2H3 cells. To investigate the anti-allergic effects of AFE or F-AFE, we examined the release of ß-hexosaminidase, a key biomarker of degranulation during an allergic reaction, and the production of pro-inflammatory mediators, such as tumor necrosis factor-α (TNF-α) and prostaglandin E2 (PGE2) in the cells treated with or without the above-mentioned extracts. AFE weakly inhibited the release of ß-hexosaminidase, whereas F-AFE significantly suppressed the release of ß-hexosaminidase in a dose-dependent manner. Consistently, F-AFE suppressed the production of TNF-α and PGE2 in a dose-dependent manner. F-AFE exerted an inhibitory effect on the production of ß-hexosaminidase, TNF-α and PGE2 with an IC50 value of 30.73, 46.96 and 36.27 µg/ml, respectively. Furthermore, F-AFE inhibited the phosphorylation of Lyn, Fyn and Syk, which are involved in the FcεRI signaling pathway, that of phosphoinositide phospholipase C (PLC)γ1/2 and protein kinase C (PKC)δ, which are associated with the degranulation process, as well as that of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase 1/2 (JNK), p38 and Akt, which are associated with cytokine expression. In the late phase, F-AFE partially suppressed the phosphorylation of cytosolic phospholipase A2 (cPLA2), but not the expression of cyclooxygenase (COX)-2. To compare and identify the major components of the two extracts, we used high-performance liquid chromatography. The levels of arctigenin, one of the major compounds, were elevated 6-fold in F-AFE compared with AFE, whereas the levels of arctiin, an arctigenin glycoside, were decreased in F-AFE by approximately 57.40%. These results suggest that arctigenin plays an important role in the anti-allergic effects of F-AFE. Taken together, F-AFE containing anti-allergic phytochemicals, including arctigenin, inhibited the activation of the FcεRI receptor induced by the antigen­IgE complex. Such effects may provide further information for the development of a phytomedicine for allergic diseases.


Subject(s)
Hypersensitivity/drug therapy , Immunoglobulin E/biosynthesis , Plant Extracts/administration & dosage , Receptors, IgE/biosynthesis , Arctium/chemistry , Fermentation , Fruit/chemistry , Gene Expression Regulation/drug effects , Humans , Hypersensitivity/immunology , Hypersensitivity/pathology , Immunoglobulin E/immunology , Intracellular Signaling Peptides and Proteins/biosynthesis , Plant Extracts/chemistry , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-fyn/biosynthesis , Receptors, IgE/immunology , Signal Transduction/drug effects , Syk Kinase , Tumor Necrosis Factor-alpha , src-Family Kinases/biosynthesis
5.
Radiat Oncol ; 10: 131, 2015 Jun 13.
Article in English | MEDLINE | ID: mdl-26071313

ABSTRACT

BACKGROUND: The mainstay of treatment in rectal cancer is neoadjuvant radio chemotherapy prior to surgery, in an attempt to downstage the tumour, allowing for more complete removal during surgery. In 40 % of cases however, this neoadjuvant radio chemotherapy fails to achieve tumour regression, partly due insufficient apoptosis signaling. X-linked Inhibitor of Apoptosis Protein (XIAP) is an anti-apoptotic protein that has been reported to contribute to disease progression and chemotherapy resistance. METHODS: We obtained rectal biopsy normal and matched tumour tissue from 29 rectal cancer patients with varying degrees of tumour regression, and using Western blot, examined anti-apoptotic XIAP and pro-apoptotic Smac protein levels in these tissues, with the aim to examine whether disturbed XIAP/Smac levels may be an indicator of neoadjuvant radio chemotherapy resistance. Expression of inhibitor of apoptosis proteins cIAP-1 and cIAP-2 was also examined. RESULTS: We found that levels of XIAP increased in accordance with the degree of radio chemotherapy resistance of the tissue. Levels of this protein were also significantly higher in tumour tissue, compared to matched normal tissue in highly resistant tissue. In contrast, Smac protein levels did not increase with radio chemotherapy resistance, and the protein was similarly expressed in normal and tumour tissue, indicating a shift in the balance of these proteins. Post treatment surgical resection tissue was available for 8 patients. When we compared matched tissue pre- and post- radio chemotherapy we found that XIAP levels increased significantly during treatment in both normal and tumour tissue, while Smac levels did not change. cIAP-1 and cIAP-2 levels were not differentially expressed in varying degrees of radio chemotherapy resistance, and neoadjuvant therapy did not alter expression of these proteins. CONCLUSION: These data indicate that disturbance of the XIAP/Smac balance may be a driver of radio chemotherapy resistance, and hence high levels of XIAP may be a useful indicator of neoadjuvant radio chemotherapy resistance in rectal cancer. Moreover, as XIAP levels increase with radio chemotherapy it is possible that a subset of more resistant tumour cells survive this treatment and may be resistant to further adjuvant treatment. Patients with resistant tumours highly expressing XIAP may benefit from alternative treatment strategies, such as Smac mimetics post neoadjuvant radio chemotherapy.


Subject(s)
Biomarkers, Tumor/analysis , Chemoradiotherapy , Drug Resistance, Neoplasm/physiology , Intracellular Signaling Peptides and Proteins/analysis , Mitochondrial Proteins/analysis , Neoadjuvant Therapy , Neoplasm Proteins/analysis , Radiation Tolerance/physiology , Rectal Neoplasms/chemistry , X-Linked Inhibitor of Apoptosis Protein/analysis , Adult , Aged , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , Baculoviral IAP Repeat-Containing 3 Protein , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Inhibitor of Apoptosis Proteins/analysis , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , X-Linked Inhibitor of Apoptosis Protein/genetics
6.
Oncol Rep ; 33(3): 1418-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25607111

ABSTRACT

An important challenge in nasopharyngeal carcinoma (NPC) research is to develop effective predictors of tumor recurrence following treatment to determine whether immediate adjuvant therapy is necessary. We retrospectively analyzed archived specimens collected from 45 patients with paired samples of primary NPC (pNPC) and recurrent NPC (rNPC). Clinical samples were collected from the Cancer Center Databases of the First People's Hospital of Foshan and Shantou Central Hospital (affiliates of Sun Yat-Sen University) between 2001 and 2012. Expression levels of phosphor-Stat3 (p-Stat3), signalosome complex subunit 5 (Jab1/Csn5), Akt1, C/EBP homologous protein (CHOP), Ki-67, and apoptosis were determined by immunohistochemistry in pNPC and rNPC samples from the same patients. Differences in these markers between the short-term interval to recurrence (ITR) group (ITR <18 months) and long-term ITR group (ITR ≥18 months) were further analyzed. In Cox's regression analysis, the ITR was significantly associated as an independent­negative prognostic factor for overall survival (hazard ratio, 0.211; 95% confidence interval, 0.053-0.841; P=0.027). p-Stat3 was increased in the short-term ITR group (ITR <18 months) and tended to be lower in the long-term ITR group (ITR ≥18 months). In the short-term ITR group, nuclear Akt expression was significantly increased in paired rNPC (P=0.028). In the long-term ITR group, the expression of nuclear Jab1/Csn5 (P=0.047) and assessment of apoptosis measured with TdT-mediated dUTP nick end­labeling (TUNEL) (P=0.003) was significantly increased in paired rNPC. The results suggest that differences between short- and long-term ITR may predict outcome in rNPC. Furthermore, the overexpression of Jab1/Csn5 and Akt may contribute to the carcinogenesis of rNPC, and Akt seems to promote the progression of short-term ITR. Intra-individual changes of Jab1/Csn5, Akt, and TUNEL may help to identify short-term ITR.


Subject(s)
Biomarkers, Tumor/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Apoptosis/physiology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , COP9 Signalosome Complex , Carcinoma , Female , Humans , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Ki-67 Antigen/biosynthesis , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Retrospective Studies , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/metabolism , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/metabolism
7.
Redox Biol ; 4: 48-59, 2015.
Article in English | MEDLINE | ID: mdl-25498967

ABSTRACT

In recent years, numerous studies have demonstrated the health benefits of polyphenols. A major portion of polyphenols in western diet are derived from coffee, which is one of the most consumed beverages in the world. It has been shown that many polyphenols gain their beneficial properties (e.g. cancer prevention) through the activation of the Nrf2/Keap1 pathway as well as their direct antioxidant activity. However, activation of Nrf2 in cancer cells might lead to resistance towards therapy through induction of phase II enzymes. In the present work we hypothesize that caffeic acid (CA), a coffee polyphenol, might act as an electrophile in addition to its nucleophilic properties and is capable of inducing the Nrf2/EpRE pathway in cancer cells. The results indicate that CA induces Nrf2 translocation into the nucleus and consequently its transcription. It has been demonstrated that generated hydrogen peroxide is involved in the induction process. It has also been found that this process is induced predominantly via the double bond in CA (Michael acceptor). However, surprisingly the presence of both nucleophilic and electrophilic moieties in CA resulted in a synergetic activation of Nrf2 and phase II enzymes. We also found that CA possesses a dual activity, although inducing GSTP1 and GSR, it inhibiting their enzymatic activity. In conclusion, the mechanism of induction of Nrf2 pathway and phase II enzymes by CA has been elucidated. The electrophilic moiety in CA is essential for the oxidation of the Keap1 protein. It should be noted that while the nucleophilic moiety (the catechol/quinone moiety) can provide scavenging ability, it cannot contribute directly to Nrf2 induction. It was found that this process may be induced by H2O2 produced by the catechol group. On the whole, it appears that CA might play a major role in the cancer cells by enhancing their resistance to treatment.


Subject(s)
Caffeic Acids/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Ovarian Neoplasms/metabolism , Antioxidants/chemistry , Antioxidants/metabolism , Caffeic Acids/chemistry , Catechols/metabolism , Cell Line, Tumor , Coffee/chemistry , Female , Humans , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/biosynthesis , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/biosynthesis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Oxidative Stress/drug effects , Polyphenols/chemistry , Polyphenols/metabolism , Signal Transduction/drug effects
8.
Oncol Rep ; 32(6): 2635-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25333415

ABSTRACT

Gastric cancer is a common malignancy with a poor prognosis. ß-elemene is a broad-spectrum anticancer drug extracted from the traditional Chinese medicinal herb Curcuma wenyujin. In the present study, we investigated the anticancer effects of ß-elemene in gastric cancer cells and the potential proteins involved. Human SGC7901 and MKN45 gastric cancer cells were treated with different concentrations of ß-elemene. Cell viability, clonogenic survival and apoptotic cell death were assessed. ß-elemene inhibited viability and decreased clonogenic survival of gastric cancer cells in a dose-dependent manner. Apoptosis induction contributed to the anticancer effects. We then employed a proteomic method, isobaric tags for relative and absolute quantitation (iTRAQ), to detect the proteins altered by ß-elemene. In total, 147 upregulated proteins and 86 downregulated proteins were identified in response to ß-elemene treatment in SGC7901 gastric cancer cells. Among them, expression of p21-activated protein kinase­interacting protein 1 (PAK1IP1), Bcl-2-associated transcription factor 1 (BTF) and topoisomerase 2-α (TOPIIα) were validated by western blot analyses and the trends were consistent with iTRAQ results. Top pathways involved in ß-elemene treatment in SGC7901 gastric cancer cells included ribosome signaling, peroxisome proliferator-activated receptors (PPARs) signaling pathway, regulation of actin cytoskeleton, phagosome, biosynthesis and metabolism of some amino acids. Collectively, our results suggest a promising therapeutic role of ß-elemene in gastric cancer. The differentially expressed proteins provide further insight into the potential mechanisms involved in gastric cancer treatment using ß-elemene.


Subject(s)
Neoplasm Proteins/biosynthesis , Proteomics , Sesquiterpenes/administration & dosage , Stomach Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
9.
J Neuroendocrinol ; 26(8): 497-509, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24943269

ABSTRACT

Notch signalling has a key role in cell fate specification in developing brains; however, recent studies have shown that Notch signalling also participates in the regulation of synaptic plasticity in adult brains. In the present study, we examined the expression of Notch3 and Delta-like ligand 4 (DLL4) in the hypothalamic-neurohypophysial system (HNS) of the adult mouse. The expression of DLL4 was higher in the supraoptic nucleus (SON) and paraventricular nucleus (PVN) compared to adjacent hypothalamic regions. Double-labelling immunohistochemistry using vesicular GABA transporter and glutamate transporter revealed that DLL4 was localised at a subpopulation of excitatory and inhibitory axonal boutons against somatodendrites of arginine vasopressin (AVP)- and oxytocin (OXT)-containing magnocellular neurones. In the neurohypophysis (NH), the expression of DLL4 was seen at OXT- but not AVP-containing axonal terminals. The expression of Notch3 was seen at somatodendrites of AVP- and OXT-containing magnocellular neurones in the SON and PVN and at pituicytes in the NH. Chronic physiological stimulation by salt loading, which remarkably enhances the release of AVP and OXT, decreased the number of DLL4-immunoreactive axonal boutons in the SON and PVN. Moreover, chronic and acute osmotic stimulation promoted proteolytic cleavage of Notch3 to yield the intracellular fragments of Notch3 in the HNS. Thus, the present study demonstrates activity-dependent reduction of DLL4 expression and proteolytic cleavage of Notch3 in the HNS, suggesting that Notch signalling possibly participates in synaptic interaction in the hypothalamic nuclei and neuroglial interaction in the NH.


Subject(s)
Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland, Posterior/metabolism , Receptors, Notch/metabolism , Signal Transduction , Supraoptic Nucleus/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Neuroglia/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Presynaptic Terminals/metabolism , Proteolysis/drug effects , Receptor, Notch3 , Receptors, Notch/biosynthesis , Sodium Chloride/pharmacology , Supraoptic Nucleus/drug effects
10.
Peptides ; 57: 122-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24874707

ABSTRACT

Large populations of cells synthesizing the neuropeptide orexin (OX) exist in the caudal hypothalamus of all species examined and are implicated in physiological and behavioral processes including arousal, stress, anxiety and depression, reproduction, and goal-directed behaviors. Hypothalamic OX expression is sexually dimorphic in different directions in laboratory rats (F>M) and mice (M>F), suggesting different roles in male and female physiology and behavior that are species-specific. We here examined if the number of hypothalamic cells immunoreactive for orexin A (OXA) differs between male and female prairie voles (Microtus ochrogaster), a socially monogamous species that pairbonds after mating and in which both sexes care for offspring, and if reproductive experience influences their number of OXA-immunoreactive (OXA-ir) cells. It was found that the total number of OXA-ir cells did not differ between the sexes, but females had more OXA-ir cells than males in anterior levels of the caudal hypothalamus, while males had more OXA-ir cells posteriorly. Sexually experienced females sacrificed 12 days after the birth of their first litter, or one day after birth of a second litter, had more OXA-ir cells in anterior levels but not posterior levels of the caudal hypothalamus compared to females housed with a brother (incest avoidance prevents sibling mating). Male prairie voles showed no effect of reproductive experience but showed an unexpected effect of cohabitation duration regardless of mating. The sex difference in the distribution of OXA-ir cells, and their increased number in anterior levels of the caudal hypothalamus of reproductively experienced female prairie voles, may reflect a sex-specific mechanism involved in pairbonding, parenting, or lactation in this species.


Subject(s)
Hypothalamus/cytology , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptides/biosynthesis , Sex Characteristics , Sexual Behavior, Animal/physiology , Animals , Arvicolinae , Female , Gene Expression Regulation , Hypothalamus/metabolism , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Neuropeptides/genetics , Orexins , Rats , Reproduction/genetics , Reproduction/physiology
11.
J Mol Endocrinol ; 52(3): T119-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532655

ABSTRACT

Neuropeptides possessing the Arg-Phe-NH2 (RFamide) motif at their C-termini (designated as RFamide peptides) have been characterized in a variety of animals. Among these, neuropeptide 26RFa (also termed QRFP) is the latest member of the RFamide peptide family to be discovered in the hypothalamus of vertebrates. The neuropeptide 26RFa/QRFP is a 26-amino acid residue peptide that was originally identified in the frog brain. It has been shown to exert orexigenic activity in mammals and to be a ligand for the previously identified orphan G protein-coupled receptor, GPR103 (QRFPR). The cDNAs encoding 26RFa/QRFP and QRFPR have now been characterized in representative species of mammals, birds, and fish. Functional studies have shown that, in mammals, the 26RFa/QRFP-QRFPR system may regulate various functions, including food intake, energy homeostasis, bone formation, pituitary hormone secretion, steroidogenesis, nociceptive transmission, and blood pressure. Several biological actions have also been reported in birds and fish. This review summarizes the current state of identification, localization, and understanding of the functions of 26RFaQRFP and its cognate receptor, QRFPR, in vertebrates.


Subject(s)
Evolution, Molecular , Neuropeptides/genetics , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence , Animals , Blood Pressure/genetics , Bone Development/genetics , Eating/genetics , Energy Metabolism/genetics , Humans , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/biosynthesis , Molecular Sequence Data , Neuropeptides/biosynthesis , Nociceptive Pain/genetics , Orexins , Sequence Alignment
12.
BMC Musculoskelet Disord ; 15: 56, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24568330

ABSTRACT

BACKGROUND: Hyperbaric oxygenation was shown to increase bone healing in a rabbit model. However, little is known about the regulatory factors and molecular mechanism involved.We hypothesized that the effect of hyperbaric oxygen (HBO) on bone formation is mediated via increases in the osteogenic differentiation of mesenchymal stem cells (MSCs) which are regulated by Wnt signaling. METHODS: The phenotypic characterization of the MSCs was analyzed by flow cytometric analysis. To investigate the effects of HBO on Wnt signaling and osteogenic differentiation of MSCs, mRNA and protein levels of Wnt3a, beta-catenin, GSK-3beta, Runx 2, as well as alkaline phosphatase activity, calcium deposition, and the intensity of von Kossa staining were analyzed after HBO treatment. To investigate the effects of HBO on Wnt processing and secretion, the expression of Wntless and vacuolar ATPases were quantified after HBO treatment. RESULTS: Cells expressed MSC markers such as CD105, CD146, and STRO-1. The mRNA and protein levels of Wnt3a, ß-catenin, and Runx 2 were up-regulated, while GSK-3ß was down-regulated after HBO treatment. Western blot analysis showed an increased ß-catenin translocation with a subsequent stimulation of the expression of target genes after HBO treatment. The above observation was confirmed by small interfering (si)RNA treatment. HBO significantly increased alkaline phosphatase activity, calcium deposition, and the intensity of von Kossa staining of osteogenically differentiated MSCs. We further showed that HBO treatment increased the expression of Wntless, a retromer trafficking protein, and vacuolar ATPases to stimulate Wnt processing and secretion, and the effect was confirmed by siRNA treatment. CONCLUSIONS: HBO treatment increased osteogenic differentiation of MSCs via regulating Wnt processing, secretion, and signaling.


Subject(s)
Bone Marrow Cells/drug effects , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Oxygen/pharmacology , Wnt Signaling Pathway/drug effects , Adult , Aged , Biomarkers , Bone Marrow Cells/metabolism , Cells, Cultured , Female , Humans , Hyperbaric Oxygenation , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Up-Regulation , Vacuolar Proton-Translocating ATPases/biosynthesis , Vacuolar Proton-Translocating ATPases/genetics , Wnt Signaling Pathway/physiology
13.
Tissue Cell ; 45(6): 452-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24138942

ABSTRACT

Neuropeptide Y (NPY) and orexin are neuropeptides involved in the regulation of feeding in vertebrates. In this study we determined the NPY and orexin mRNA tissue expression and their immunoreactivity distribution in both preoptic area and hypothalamus, regions involved in the regulation of feeding behavior. Both peptides presented a wide expression in all tissues examined. The NPY-immunoreactive (ir) cells were localized in the ventral nucleus posterioris periventricularis (NPPv) and numerous ir-NPY fibers were found in the nucleus lateralis tuberis (NLT), the nucleus recess lateralis (NRL) and the neurohypophysis. Ir-orexin cells were observed in the NPPv, dorsal NLT, ventral NLT, lateral NLT (NLTl) and the lateral NRL. Ir-orexin fibers were widespread distributed along all the hypothalamus, especially in the NLTl. Additionally, we observed the presence of ir-orexin immunostaining in adenohypophyseal cells, especially in somatotroph cells and the presence of a few ir-orexin-A fibers in the neurohypophysis. In conclusion, both peptides have an ubiquitous mRNA tissue expression and are similarly distributed in the hypothalamus and preoptic area of Cichlasoma dimerus. The presence of ir-orexin in adenohypohyseal cells and the presence of ir-orexin and NPY fibers in the neurohypophysis suggest that both peptides may play an important neuroendocrine role in anterior pituitary.


Subject(s)
Cichlids/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Animals , Cichlids/genetics , Orexins , Preoptic Area/metabolism
14.
J Biol Chem ; 288(44): 31423-36, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045941

ABSTRACT

Signaling lymphocytic activation molecule-associated protein (SAP) is an Src homology 2 domain-only adaptor involved in multiple immune cell functions. It has also been linked to immunodeficiencies and autoimmune diseases, such as systemic lupus erythematosus. Here, we examined the role and mechanism of action of SAP in autoimmunity using a mouse model of autoimmune arthritis, collagen-induced arthritis (CIA). We found that SAP was essential for development of CIA in response to collagen immunization. It was also required for production of collagen-specific antibodies, which play a key role in disease pathogenesis. These effects required SAP expression in T cells, not in B cells. In mice immunized with a high dose of collagen, the activity of SAP was nearly independent of its ability to bind the protein tyrosine kinase Fyn and correlated with the capacity of SAP to promote full differentiation of follicular T helper (TFH) cells. However, with a lower dose of collagen, the role of SAP was more dependent on Fyn binding, suggesting that additional mechanisms other than TFH cell differentiation were involved. Further studies suggested that this might be due to a role of the SAP-Fyn interaction in natural killer T cell development through the ability of SAP-Fyn to promote Vav-1 activation. We also found that removal of SAP expression during progression of CIA attenuated disease severity. However, it had no effect on disease when CIA was clinically established. Together, these results indicate that SAP plays an essential role in CIA because of Fyn-independent and Fyn-dependent effects on TFH cells and, possibly, other T cell types.


Subject(s)
Arthritis, Experimental/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn/metabolism , T-Lymphocytes/metabolism , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes , Cell Line , Collagen/toxicity , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Proto-Oncogene Proteins c-vav/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Sleep ; 36(8): 1129-38, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23904672

ABSTRACT

STUDY OBJECTIVES: Narcolepsy is caused by selective loss of the orexin/hypocretin-producing neurons of the hypothalamus. For patients with narcolepsy, chronic sleepiness is often the most disabling symptom, but current therapies rarely normalize alertness and do not address the underlying orexin deficiency. We hypothesized that the sleepiness of narcolepsy would substantially improve if orexin signaling were restored in specific brain regions at appropriate times of day. DESIGN: We used gene therapy to restore orexin signaling in a mouse model of narcolepsy. In these Atx mice, expression of a toxic protein (ataxin-3) selectively kills the orexin neurons. INTERVENTIONS: To induce ectopic expression of the orexin neuropeptides, we microinjected an adeno-associated viral vector coding for prepro-orexin plus a red fluorescence protein (AAV-orexin) into the mediobasal hypothalamus of Atx and wild-type mice. Control mice received an AAV coding only for red fluorescence protein. Two weeks later, we recorded sleep/wake behavior, locomotor activity, and body temperature and examined the patterns of orexin expression. MEASUREMENTS AND RESULTS: Atx mice rescued with AAV-orexin produced long bouts of wakefulness and had a normal diurnal pattern of arousal, with the longest bouts of wake and the highest amounts of locomotor activity in the first hours of the night. In addition, AAV-orexin improved the timing of rapid eye movement sleep and the consolidation of nonrapid eye movement sleep in Atx mice. CONCLUSIONS: These substantial improvements in sleepiness and other symptoms of narcolepsy demonstrate the effectiveness of orexin gene therapy in a mouse model of narcolepsy. Additional work is needed to optimize this approach, but in time, AAV-orexin could become a useful therapeutic option for patients with narcolepsy.


Subject(s)
Genetic Therapy/methods , Intracellular Signaling Peptides and Proteins/genetics , Narcolepsy/therapy , Neuropeptides/genetics , Wakefulness/genetics , Animals , Ataxin-3 , Disease Models, Animal , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/physiology , Male , Mice , Mice, Transgenic/genetics , Mice, Transgenic/physiology , Narcolepsy/genetics , Neuropeptides/biosynthesis , Neuropeptides/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Orexins , Polysomnography , Sleep/genetics , Sleep/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Wakefulness/physiology
16.
Oncol Rep ; 30(1): 292-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23633038

ABSTRACT

Plant-derived active constituents and their semi-synthetic or synthetic analogs have served as major sources of anticancer drugs. 20(S)-protopanaxadiol (PPD) is a metabolite of ginseng saponin of both American ginseng (Panax quinquefolius L.) and Asian ginseng (Panax ginseng C.A. Meyer). We previously demonstrated that ginsenoside Rg3, a glucoside precursor of PPD, exhibits anti-proliferative effects on HCT116 cells and reduces tumor size in a xenograft model. Our subsequent study indicated that PPD has more potent antitumor activity than that of Rg3 in vitro although the mechanism underlying the anticancer activity of PPD remains to be defined. Here, we investigated the mechanism underlying the anticancer activity of PPD in human cancer cells in vitro and in vivo. PPD was shown to inhibit growth and induce cell cycle arrest in HCT116 cells. The in vivo studies indicate that PPD inhibits xenograft tumor growth in athymic nude mice bearing HCT116 cells. The xenograft tumor size was significantly reduced when the animals were treated with PPD (30 mg/kg body weight) for 3 weeks. When the expression of previously identified Rg3 targets, A kinase (PRKA) anchor protein 8 (AKAP8L) and phosphatidylinositol transfer protein α (PITPNA), was analyzed, PPD was shown to inhibit the expression of PITPNA while upregulating AKAP8L expression in HCT116 cells. Pathway-specific reporter assays indicated that PPD effectively suppressed the NF-κB, JNK and MAPK/ERK signaling pathways. Taken together, our results suggest that the anticancer activity of PPD in colon cancer cells may be mediated through targeting NF-κB, JNK and MAPK/ERK signaling pathways, although the detailed mechanisms underlying the anticancer mode of PPD action need to be fully elucidated.


Subject(s)
MAP Kinase Signaling System/drug effects , Neoplasms/drug therapy , Sapogenins/pharmacology , Sapogenins/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/biosynthesis , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Nude , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Neoplasm Transplantation , Nuclear Proteins/biosynthesis , Panax , Phospholipid Transfer Proteins/biosynthesis , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Signal Transduction/drug effects
17.
J Physiol Pharmacol ; 64(1): 55-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23568972

ABSTRACT

Orexins A and B are hypothalamic peptides engaged in a variety of physiological functions related to the control of energy homeostasis, sleep and wakefulness. The presence of orexin receptors in the tissues of the hypothalamus-pituitary-gonadal axis indicates that these hormones are also involved in the control of the reproductive system. The aim of this study was to compare the expression levels of prepro-orexin (a precursor of orexins A and B) mRNA in the porcine hypothalamic structures involved in reproductive processes - mediobasal hypothalamus (MBH), preoptic area (POA) and stalk median eminence (SME), during four stages (days 2-3, 10-12, 14-16, 17-19) of the oestrous cycle. In MBH, lower concentrations of PPO mRNA were observed on days 2-3 than in the remaining stages. In POA, the highest mRNA expression of PPO was noted on days 17-19. In SME, the highest concentrations of PPO was observed on days 2-3, and the lowest on days 14-16. We also investigated the intensity of OXA and OXB immunoreactivity and detected both peptides in all examined structures. In MBH, signal intensity for OXA was highest on days 14-16 and lowest on days 17-19. The highest levels of immunoreactivity were noted on days 2-3 and 10-12 in POA, and in SME additionally on days 17-19. OXB immunoreactivity in hypothalamic tissues also changed during the cycle, and the highest signal intensity was reported on days 10-12 in MBH, on days 14-16 in POA, and on days 14-16 and 17-19 in SME. The results of our study indicate that orexins A and B are produced in the porcine hypothalamus and that their concentrations vary subject to the pig's hormonal status. Our findings also suggest that orexins may affect reproductive functions at the highest level of the hypothalamus-pituitary-gonadal axis.


Subject(s)
Estrous Cycle/genetics , Estrous Cycle/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptides/biosynthesis , Animals , Female , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Median Eminence/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Orexins , Preoptic Area/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Swine/genetics , Swine/metabolism
18.
Proc Natl Acad Sci U S A ; 110(18): 7136-41, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23596212

ABSTRACT

Biogenesis of iron-sulfur cluster proteins is a highly regulated process that requires complex protein machineries. In the cytosolic iron-sulfur protein assembly machinery, two human key proteins--NADPH-dependent diflavin oxidoreductase 1 (Ndor1) and anamorsin--form a stable complex in vivo that was proposed to provide electrons for assembling cytosolic iron-sulfur cluster proteins. The Ndor1-anamorsin interaction was also suggested to be implicated in the regulation of cell survival/death mechanisms. In the present work we unravel the molecular basis of recognition between Ndor1 and anamorsin and of the electron transfer process. This is based on the structural characterization of the two partner proteins, the investigation of the electron transfer process, and the identification of those protein regions involved in complex formation and those involved in electron transfer. We found that an unstructured region of anamorsin is essential for the formation of a specific and stable protein complex with Ndor1, whereas the C-terminal region of anamorsin, containing the [2Fe-2S] redox center, transiently interacts through complementary charged residues with the FMN-binding site region of Ndor1 to perform electron transfer. Our results propose a molecular model of the electron transfer process that is crucial for understanding the functional role of this interaction in human cells.


Subject(s)
Flavoproteins/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Iron-Sulfur Proteins/biosynthesis , Oxidoreductases/biosynthesis , Protein Biosynthesis , Electron Transport , Flavin Mononucleotide/metabolism , Flavoproteins/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Iron-Sulfur Proteins/chemistry , Models, Biological , Models, Molecular , Oxidoreductases/chemistry , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary
19.
Neuropeptides ; 47(3): 139-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23490004

ABSTRACT

The hypothalamic neurochemicals neuropeptide Y (NPY), orexin-A (ORX), and oxytocin (OXY) exert glucoregulatory effects upon intracerebral administration, findings that support their potential function within neural pathways that maintain glucostasis. Current understanding of how these neurotransmitter systems respond to the diabetes mellitus complication, insulin-induced hypoglycemia, is limited to knowledge of neuropeptide gene transcriptional reactivity. We investigated the hypothesis that hypoglycemia elicits hypothalamic site-specific alterations in levels of these neurochemicals, and that adjustments in local neurotransmitter availability may be regulated by catecholaminergic (CA) input from the caudal dorsomedial hindbrain. The arcuate (ARH) and paraventricular (PVH) hypothalamic nuclei and lateral hypothalamic area (LHA) were each microdissected from adult male rats pretreated by caudal fourth ventricular administration of the selective CA neurotoxin, 6-hydroxydopamine (6-OHDA), or vehicle prior to insulin (INS)-induced hypoglycemia. Hypoglycemia stimulated ARH NPY gene expression and NPY accumulation in the ARH and LHA, but not PVH. 6-OHDA pretreatment did not modify the positive NPY mRNA response to INS, but blunted hypoglycemic augmentation of ARH and LHA NPY content while increasing PVH NPY levels in response to hypoglycemia. INS-treated rats exhibited diminished LHA ORX gene expression and increased [ARH; LHA] or decreased [PVH] tissue ORX protein levels. 6-OHDA+INS animals showed a comparable decline in ORX transcripts, but attenuated augmentation of ARH and LHA ORX content and elevated PVH ORX levels. OT mRNA and protein were respectively decreased or unchanged during hypoglycemia, responses that were uninfluenced by hindbrain CA nerve cell destruction. These results illustrate divergent adjustments in glucoregulatory neurotransmitter gene expression and site-specific protein accumulation in the hypothalamus during hypoglycemia. Evidence that 6-OHDA pretreatment does not modify NPY or ORX transcriptional reactivity to hypoglycemia, but alters hypoglycemic patterns of NPY and ORX accretion implicates dorsomedial hindbrain CA neurons in regulation of translation/post-translational processing and site-specific availability of these neurotransmitters in the hypothalamus during hypoglycemia.


Subject(s)
Catecholamines/metabolism , Glucose/metabolism , Hypoglycemia/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Oxytocin/biosynthesis , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Hydroxydopamines/pharmacology , Hypoglycemia/chemically induced , Hypoglycemic Agents , Immunohistochemistry , Insulin , Intracellular Signaling Peptides and Proteins/genetics , Male , Neuropeptide Y/genetics , Neuropeptides/genetics , Neurotoxins/metabolism , Neurotransmitter Agents/metabolism , Orexins , Oxytocin/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sympathectomy, Chemical
20.
Psychoneuroendocrinology ; 38(4): 560-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22917622

ABSTRACT

The orexin neuropeptide system regulates wakefulness and contributes to physiological and behavioral stress responses. Moreover, a role for orexins in modulating hypothalamus-pituitary-adrenal (HPA) axis activity has been proposed. Brain penetrating dual orexin receptor (OXR) antagonists such as almorexant decrease vigilance and have emerged as a novel therapeutic class for the treatment of insomnia. Almorexant was used here as a pharmacological tool to examine the role of endogenous orexin signaling in HPA axis endocrine function under natural conditions. After confirming the expression of prepro-orexin and OXR-1 and OXR-2 mRNA in hypothalamus, pituitary and adrenal glands, the effects of systemic almorexant were investigated on peripheral HPA axis hormone release in the rat under baseline, stress and pharmacological challenge conditions. Almorexant did not alter basal or stress-induced corticosterone release despite affecting wake and sleep stages (detected by radiotelemetric electroencephalography/electromyography) during the stress exposure. Moreover, almorexant did not affect the release of adrenocorticotropin (ACTH) and corticosterone at different time points along the diurnal rhythm, nor corticotrophin-releasing hormone (CRH)- and ACTH-stimulated neuroendocrine responses, measured in vivo under stress-free conditions. These results illustrate that dual OXR antagonists, despite modulating stress-induced wakefulness, do not interfere with endocrine HPA axis function in the rat. They converge to suggest that endogenous orexin signaling plays a minor role in stress hormone release under basal conditions and under challenge.


Subject(s)
Acetamides/pharmacology , Adrenal Glands/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Isoquinolines/pharmacology , Neuropeptides/physiology , Orexin Receptor Antagonists , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/blood , Animals , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Corticosterone/blood , Corticotropin-Releasing Hormone/pharmacology , Hypnotics and Sedatives/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Neuropeptides/biosynthesis , Orexin Receptors/biosynthesis , Orexins , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Restraint, Physical/physiology , Sleep Stages/drug effects , Wakefulness/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL