Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 516(3): 851-857, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31266633

ABSTRACT

A number of homeobox genes are implicated in the malignancy of various cancers. Here, we investigated the role of the homeobox gene SIX4 in non-small-cell lung cancer (NSCLC). The sine oculis homeobox (SIX4) gene was found to be highly expressed at both mRNA and protein levels in NSCLC tumor tissues as compared with matching normal counterparts. In this study, the SIX4 gene of two human NSCLC cell lines (A549 and PC9) was overexpressed or silenced using the lentiviral system. We evaluated the malignancy-associated phenotype of transfected cells, which demonstrated that exogenous expression of the SIX4 gene greatly enhanced the proliferation, migration, and invasion of NSCLC cells. The opposite was true in the SIX4-silenced cells. Transcriptomic profiling analysis revealed that the SIX4 gene modulated the expression of hundreds of downstream target genes in a cell context-dependent manner. Most notably, the SIX4 gene controls the expression of crucial genes with evidently oncogenic function. We conclude that SIX4 plays an oncogenic role and may be potentially utilized as a diagnostic and therapeutic marker for NSCLC.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Trans-Activators/genetics , A549 Cells , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Profiling , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Homeodomain Proteins/agonists , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trans-Activators/agonists , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
2.
Toxicol Lett ; 294: 11-19, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29746904

ABSTRACT

This study aimed to investigate the potential roles of TG-interacting factor (TGIF) in benzo(a)pyrene (BaP)-induced migration, invasion, and metastasis of lung adenocarcinoma cells. Cells were treated with different concentrations of BaP. MTT assays were used to measure cell proliferation. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunoblots were applied to measure the TGIF expression. A dual-luciferase reporter gene assay was performed to assess the effects of BaP on TGIF promoter-driven reporter gene expression. Wound-healing, transwell, and tail vein metastasis assays were performed to evaluate migratory, invasive, and metastatic capacity. Our results showed that BaP treatment increased the expression of TGIF mRNA and protein. Additionally, BaP treatment enhanced TGIF promoter-driven reporter gene expression. We observed that BaP treatment promoted the migration, invasion, and metastasis of H157 cells, which could be blocked by silencing TGIF. The expression of TGIF mRNA was significantly higher in metastatic lung adenocarcinoma samples than in non-metastatic lung adenocarcinoma samples, and higher levels of TGIF mRNA expression were observed in metastatic lung adenocarcinoma samples from patients with a smoking history than in those from patients with a non-smoking history. Our findings suggest that BaP treatment promotes the migration, invasion, and metastasis of human lung adenocarcinoma cells by upregulating TGIF.


Subject(s)
Adenocarcinoma/chemically induced , Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/agonists , Lung Neoplasms/chemically induced , Promoter Regions, Genetic/drug effects , Repressor Proteins/agonists , Up-Regulation/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adenocarcinoma of Lung , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Genes, Reporter/drug effects , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , RNA Interference , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Smoking/adverse effects , Tumor Burden/drug effects
3.
Free Radic Biol Med ; 110: 280-290, 2017 09.
Article in English | MEDLINE | ID: mdl-28666851

ABSTRACT

Cancer recurrence, which is frequently accompanied by chemotherapy, has been a challenge in cancer treatment. This study was carried out to examine the potential applications of the reactive oxygen species (ROS)-producing cold atmospheric plasma (CAP) to overcome the cancer cells' drug resistance, which has been emerging as an alternative therapeutic tool for cancer. For this, we developed a tamoxifen (Tam)-resistant MCF-7 (MCF-7/TamR) breast cancer cell model and examined the effect of CAP on the recovery of Tam sensitivity at the cellular and molecular level. The ROS level was increased 1.9-fold in CAP-treated MCF-7/TamR cells compared to the non-treated cell. CAP was proven to restore sensitivity by up to 50% for MCF-7/TamR cells against Tam after CAP treatment. The comparison of genome-wide expression between the acquisition of Tam resistance and CAP treatment identified 20 genes that commonly showed significant expression changes. Notably, all the genes except two have been oppositely dysregulated in the two cellular statuses, and the majority of them are known to contribute to the acquisition of Tam resistance. The protein expression of selected genes, MX1 and HOXC6, was recovered to that of their parental cell by CAP. Furthermore, the dysregulation of MX1 and HOXC6 in MCF-7/TamR alleviated the drug sensitivity recovery effect of CAP. Taken together, CAP inhibited the growth of Tam-resistant MCF-7 cancer cells and reset it to the Tam-sensitive status by restoring the expression of drug resistance-related genes. These findings may lend credence to CAP as an alternative or complementary tool in the treatment or prevention of Tam-resistant cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Plasma Gases/pharmacology , Tamoxifen/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Genome-Wide Association Study , Homeodomain Proteins/agonists , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MCF-7 Cells , Myxovirus Resistance Proteins/agonists , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Biosci Biotechnol Biochem ; 80(7): 1425-32, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26940726

ABSTRACT

Bone homeostasis is maintained by balancing bone formation and bone resorption, but an imbalance between them is associated with various bone-related diseases such as osteoporosis and rheumatoid arthritis. We found that 5,6-dehydrokawain (DK) and dihydro-5,6-dehydrokawain (DDK), which were isolated as promising compounds from Alpinia zerumbet rhizomes, promote differentiation of osteoblastic MC3T3-E1 cells. DK and DDK increased the alkaline phosphatase activity and matrix mineralization of MC3T3-E1 cells. DK exerts larger effects than DDK. The gene expression of runt-related transcription factor 2 and osterix, which are essential transcription factors in the early period of osteoblast differentiation, was significantly increased by DK treatment. The mRNA level of distal-less homeobox 5 was also enhanced by DK treatment, and DK activated the p38 mitogen-activated protein kinase pathway. Therefore, DK may have clinical potential for preventing osteoporosis, and could be considered as a potential anabolic therapeutic agent.


Subject(s)
Alpinia/chemistry , Cell Differentiation/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , Pyrones/pharmacology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Core Binding Factor Alpha 1 Subunit/agonists , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation , Homeodomain Proteins/agonists , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Plant Extracts/chemistry , Pyrones/isolation & purification , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhizome/chemistry , Sp7 Transcription Factor , Transcription Factors/agonists , Transcription Factors/genetics , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Molecules ; 19(4): 4654-68, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24739928

ABSTRACT

A physio-pathological feature of diabetes mellitus is a significant reduction of ß-pancreatic cells. The growth, differentiation and function maintenance of these cells is directed by transcription factors. Nkx6.1 is a key transcription factor for the differentiation, neogenesis and maintenance of ß-pancreatic cells. We reported that silymarin restores normal morphology and endocrine function of damaged pancreatic tissue after alloxan-induced diabetes mellitus in rats. The aim of this study was to analyze the effect of silymarin on Nkx6.1 transcription factor expression and its consequence in ß cells neogenesis. Sixty male Wistar rats were partially pancreatectomized and divided into twelve groups. Six groups were treated with silymarin (200 mg/Kg p.o) for periods of 3, 7, 14, 21, 42 and 63 days. Additionally, an unpancreatectomized control group was used. Nkx6.1 and insulin gene expression were assessed by RT-PCR assay in total pancreatic RNA. ß-Cell neogenesis was determined by immunoperoxidase assay. Silymarin treated group showed an increase of Nkx6.1 and insulin genic expression. In this group, there was an increment of ß-cell neogenesis in comparison to pancreatectomized untreated group. Silymarin treatment produced a rise in serum insulin and serum glucose normalization. These results suggest that silymarin may improve the reduction of ß pancreatic cells observed in diabetes mellitus.


Subject(s)
Homeodomain Proteins/agonists , Insulin-Secreting Cells/drug effects , Insulin/agonists , Pancreatectomy , Protective Agents/pharmacology , Silymarin/pharmacology , Animals , Blood Glucose/metabolism , Cell Count , Cell Proliferation , Diabetes Mellitus , Disease Models, Animal , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Rats , Rats, Wistar
6.
PLoS One ; 9(2): e90093, 2014.
Article in English | MEDLINE | ID: mdl-24587221

ABSTRACT

AIMS/HYPOTHESIS: Incretin therapies, which are used to treat diabetic patients, cause a chronic supra-physiological increase in GLP-1 circulating levels. It is still unclear how the resulting high hormone concentrations may affect pancreatic alpha cells. The present study was designed to investigate the effects of chronic exposure to high GLP-1 levels on a cultured pancreatic alpha cell line. METHODS: α-TC1-6 cell line was cultured in the presence or absence of GLP-1 (100 nmol/l) for up to 72 h. In our model GLP-1 receptor (GLP-1R) was measured. After the cells were exposed to GLP-1 the levels of glucagon secretion were measured. Because GLP-1 acts on intracellular cAMP production, the function of GLP-1R was studied. We also investigated the effects of chronic GLP-1 exposure on the cAMP/MAPK pathway, Pax6 levels, the expression of prohormone convertases (PCs), glucagon gene (Gcg) and protein expression, glucagon and GLP-1 production. RESULTS: In our model, we were able to detect GLP-1R. After GLP-1 exposure we found a reduction in glucagon secretion. During further investigation of the function of GLP-1R, we found an activation of the cAMP/MAPK/Pax6 pathway and an increase of Gcg gene and protein expression. Furthermore we observed a significant increase in PC1/3 protein expression, GLP-1 intracellular content and GLP-1 secretion. CONCLUSIONS/INTERPRETATION: Our data indicate that the chronic exposure of pancreatic alpha cells to GLP-1 increases the ability of these cells to produce and release GLP-1. This phenomenon occurs through the stimulation of the transcription factor Pax6 and the increased expression of the protein convertase PC1/3.


Subject(s)
Eye Proteins/genetics , Glucagon-Like Peptide 1/pharmacology , Glucagon-Secreting Cells/drug effects , Homeodomain Proteins/genetics , Paired Box Transcription Factors/genetics , Proprotein Convertase 1/genetics , Repressor Proteins/genetics , Cell Line, Tumor , Cyclic AMP/metabolism , Eye Proteins/agonists , Eye Proteins/metabolism , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/agonists , Homeodomain Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/agonists , Paired Box Transcription Factors/metabolism , Proprotein Convertase 1/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Repressor Proteins/agonists , Repressor Proteins/metabolism , Signal Transduction
7.
J Cell Sci ; 127(Pt 1): 40-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24155330

ABSTRACT

Cardiac fibrosis is linked to fibroblast-to-myofibroblast phenoconversion and proliferation but the mechanisms underlying this are poorly understood. Ski is a negative regulator of TGF-ß-Smad signaling in myofibroblasts, and might redirect the myofibroblast phenotype back to fibroblasts. Meox2 could alter TGF-ß-mediated cellular processes and is repressed by Zeb2. Here, we investigated whether Ski diminishes the myofibroblast phenotype by de-repressing Meox2 expression and function through repression of Zeb2 expression. We show that expression of Meox1 and Meox2 mRNA and Meox2 protein is reduced during phenoconversion of fibroblasts to myofibroblasts. Overexpression of Meox2 shifts the myofibroblasts into fibroblasts, whereas the Meox2 DNA-binding mutant has no effect on myofibroblast phenotype. Overexpression of Ski partially restores Meox2 mRNA expression levels to those in cardiac fibroblasts. Expression of Zeb2 increased during phenoconversion and Ski overexpression reduces Zeb2 expression in first-passage myofibroblasts. Furthermore, expression of Meox2 is decreased in scar following myocardial infarction, whereas Zeb2 protein expression increases in the infarct scar. Thus Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by upregulating the expression of Meox2. This cascade might regulate cardiac myofibroblast phenotype and presents therapeutic options for treatment of cardiac fibrosis.


Subject(s)
Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation , Homeodomain Proteins/agonists , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Muscle Proteins/agonists , Muscle Proteins/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myofibroblasts/pathology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
8.
ACS Chem Biol ; 5(11): 1035-43, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20822188

ABSTRACT

Bone marrow stromal cells (BMSCs) play critical roles in repairing endothelium damage. However, the mechanisms underlying BMSC differentiation into vascular endothelial cells (VECs) is not well understood. We aimed to find new factors involved in this process by exploiting a novel chemical inducer in a gene microarray assay. We first identified a novel benzoxazine derivative (6-amino-2,3-dihydro-3-hydroxymethyl-1,4-benzoxazine; ABO) that can induce BMSC differentiation to VECs in a capillary-like tube formation assay, promote analysis of endothelial cell-specific marker expression, and facilitate uptake of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate-acetylated low-density lipoprotein (Dil-Ac-LDL). Microarray analysis of BMSCs treated with ABO for 4 h revealed changes in only a handful of genes. The only one upregulated was homeobox-containing 1 (Hmbox1) gene, whereas six genes, including IP-10 and others, were downregulated. The upregulation of Hmbox1 and downregulation of IP-10 were confirmed by RT-PCR, quantitative PCR (qPCR), and Western blot analysis. It is reported that IP-10 could suppresse EC differentiation into capillary structures. In this study ABO could not induce BMSC differentiation to VECs in the presence of IP-10. Small interfering RNA knockdown of Hmbox1 blocked ABO-induced BMSC differentiation and increased the level of IP-10 but decreased Ets-1. Thus, ABO is a novel inducer for BMSC differentiation to VECs, and Hmbox1 is a key factor in the differentiation. IP-10 and Ets-1 might be relevant targets of Hmbox1 in BMSC differentiation to VECs. These findings provide information on a novel target and a new platform for further investigating the gene control of BMSC differentiation to VECs.


Subject(s)
Benzoxazines/pharmacology , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Endothelium, Vascular/cytology , Homeodomain Proteins/physiology , Neovascularization, Physiologic/physiology , Animals , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Endothelium, Vascular/physiology , Gene Expression Regulation/drug effects , Homeodomain Proteins/agonists , Homeodomain Proteins/genetics , Male , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/physiology
9.
Exp Cell Res ; 315(18): 3163-75, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19766113

ABSTRACT

Agents inducing O(6)-methylguanine (O(6)MeG) in DNA such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) are cytotoxic and a deficiency in mismatch repair (MMR) results in lack of sensitivity to this genotoxin (termed alkylation tolerance). Here, we show that ING2, a member of the inhibitor of growth family, is required for cell death induced by MNNG. We further observe that MNNG treatment increases cellular protein levels of ING2 that is dependent on intact MMR function and that MNNG-induced ING2 localizes and associates with p73alpha in the nucleus. Suppression of ING2 by short hairpin RNA (shRNA) in MMR-proficient colorectal cancer cells decreased its sensitivity to MNNG and, in addition, abrogated MNNG-induced stabilization and acetylation of p73alpha. Interestingly, suppression of p73alpha had a greater impact on MNNG-induced cell death than ING2 leading us to conclude that ING2 regulates the cell death response, in part, through p73alpha. Inhibition of c-Abl by STI571 or suppression of c-Abl expression by shRNA blocked ING2 induction and p73alpha acetylation induced by this alkylator. Similarly, suppression of MMR (MLH1) by shRNA abrogated ING2 induction/p73alpha acetylation. Taken together, these results demonstrate that MLH1/c-Abl-dependent activation of ING2>p73alpha signaling regulates cell death triggered by MNNG and further suggests that dysregulation of this event may, in part, be responsible for alkylation tolerance observed in MMR compromised cells.


Subject(s)
Alkylating Agents/toxicity , Apoptosis , Carcinogens/toxicity , DNA Mismatch Repair , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Methylnitronitrosoguanidine/toxicity , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Benzamides , DNA-Binding Proteins/antagonists & inhibitors , HeLa Cells , Homeodomain Proteins/agonists , Humans , Imatinib Mesylate , MutL Protein Homolog 1 , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/drug effects , Oligonucleotides/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/drug effects , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Tumor Protein p73 , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/agonists , Tumor Suppressor Proteins/antagonists & inhibitors
10.
Alcohol Clin Exp Res ; 32(8): 1361-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18565153

ABSTRACT

INTRODUCTION: Prenatal alcohol exposure via maternal liquid diet consumption by C57BL/6 (B6) mice causes conspicuous midline neural tube deficit (dysraphia) and disruption of genesis and development of serotonin (5-HT) neurons in the raphe nuclei, together with brain growth retardation. The current study tested the hypothesis that concurrent treatment with either an activity-dependent neurotrophic factor (ADNF) agonist peptide [SALLRSIPA, (SAL)] or an activity-dependent neurotrophic protein (ADNP) agonist peptide [NAPVSIPQ, (NAP)] would protect against these alcohol-induced deficits in brain development. METHODS: Timed-pregnant B6 dams consumed alcohol from embryonic day 7 (E7, before the onset of neurulation) until E15. Fetuses were obtained on E15 and brain sections processed for 5-HT immunocytochemistry, for evaluation of morphologic development of the brainstem raphe and its 5-HT neurons. Additional groups were treated either with SAL or NAP daily from E7 to E15 to assess the potential protective effects of these peptides. Measures of incomplete occlusion of the ventral canal and the frequency and extent of the openings in the rhombencephalon were obtained to assess fetal dysraphia. Counts of 5-HT-immunostained neurons were also obtained in the rostral and caudal raphe. RESULTS: Prenatal alcohol exposure resulted in abnormal openings along the midline and delayed closure of ventral canal in the brainstem. This dysraphia was associated with reductions in the number of 5-HT neurons both in the rostral raphe nuclei (that gives rise to ascending 5-HT projections) and in the caudal raphe (that gives rise to the descending 5-HT projections). Concurrent treatment of the alcohol-consuming dams with SAL prevented dysraphia and protected against the alcohol-induced reductions in 5-HT neurons in both the rostral and caudal raphe. NAP was less effective in protecting against dysraphia and did not protect against 5-HT loss in the rostral raphe, but did protect against loss in the caudal raphe. CONCLUSIONS: These findings further support the potential usefulness of these peptides for therapeutic interventions in pregnancies at risk for alcohol-induced developmental deficits. Notably, the ascending 5-HT projections of the rostral raphe have profound effects in regulating forebrain development and function, and the descending 5-HT projections of the caudal raphe are critical for regulating respiration. Protection of the rostral 5-HT-system may help prevent structural and functional deficits linked to abnormal forebrain development, and protection of the caudal systems may also reduce the increased risk for sudden infant death syndrome associated with prenatal alcohol exposure.


Subject(s)
Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Homeodomain Proteins/agonists , Nerve Tissue Proteins/agonists , Neural Tube Defects/prevention & control , Peptide Fragments/therapeutic use , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Central Nervous System Depressants/pharmacology , Disease Models, Animal , Ethanol/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Neural Tube Defects/chemically induced , Neurons/drug effects , Neurons/metabolism , Neuropeptides , Oligopeptides , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/prevention & control , Prenatal Injuries/chemically induced , Prenatal Injuries/prevention & control , Respiration/drug effects , Serotonin/metabolism , Spinal Dysraphism/chemically induced , Spinal Dysraphism/prevention & control
11.
J Med Chem ; 49(23): 6652-5, 2006 Nov 16.
Article in English | MEDLINE | ID: mdl-17154495

ABSTRACT

We report the identification of substituted cis-bicyclo[3.3.0]-oct-2-enes as small molecule agonists of subfamily V orphan nuclear receptors (NR5A), liver receptor homolog-1 (LRH-1) and steroidogenic factor-1 (SF-1). Using fluorescence resonance energy transfer (FRET)-based biochemical assays, compound 5a (GSK8470) was identified as a high-affinity ligand for LRH-1 and SF-1. In liver cells, 5a increased the expression of the LRH-1 target gene small heterodimer partner (SHP). Synthesis of analogues modified at three positions led to the development of compounds with functional selectivity between LRH-1 and SF-1.


Subject(s)
Alkenes/chemical synthesis , Aniline Compounds/chemical synthesis , Bridged Bicyclo Compounds/chemical synthesis , DNA-Binding Proteins/agonists , Homeodomain Proteins/agonists , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Alkenes/chemistry , Alkenes/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Binding Sites , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacology , Cells, Cultured , Fluorescence Resonance Energy Transfer , Genes, Reporter , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Ligands , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Stereoisomerism , Steroidogenic Factor 1 , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...