Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Steroids ; 78(2): 137-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23182766

ABSTRACT

The translocator protein (18-kDa) TSPO is an ubiquitous high affinity cholesterol-binding protein reported to be present in the endothelial and smooth muscle cells of the blood vessels; its expression dramatically increased in macrophages found in atherosclerotic plaques. A domain in the carboxy-terminus of TSPO was identified and characterized as the cholesterol recognition/interaction amino acid consensus (CRAC). The ability of the CRAC domain to bind to cholesterol led us to hypothesize that this peptide could be used as an hypocholesterolemic, with potential anti-atherogenic properties, agent. We report herein the therapeutic benefit that resulted for the administration of the VLNYYVWR human CRAC sequence to guinea pigs fed with a high cholesterol diet and ApoE knock-out B6.129P2-Apoetm1Unc/J mice. CRAC treatment (3 and 30mg/kg once daily for 6 weeks) resulted in reduced circulating cholesterol levels in guinea pigs fed with 2% high cholesterol diet and ApoE knock-out B6.129P2-Apoetm1Unc/J mice. In high cholesterol fed guinea pigs, CRAC treatment administered once daily induced an increase in circulating HDL, decreased total, free and LDL cholesterol, and removed atheroma deposits in the aorta in a dose-dependent manner. The treatment also prevented the high cholesterol diet-induced increase in serum creatine kinase, total and isoforms, markers of neurological, cardiac and muscular damage. No toxicity was observed. Taken together these results support a role of TSPO in lipid homeostasis and atherosclerosis and indicate that CRAC may constitute a novel and safe treatment of hypercholesterolemia and atherosclerosis.


Subject(s)
Atherosclerosis/complications , Atherosclerosis/drug therapy , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Receptors, GABA/chemistry , Receptors, GABA/therapeutic use , Amino Acid Sequence , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/blood , Body Weight/drug effects , Cholesterol/blood , Cholesterol, Dietary/pharmacology , Creatine Kinase/blood , Guinea Pigs , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hypercholesterolemia/blood , Immunohistochemistry , Isoenzymes/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Molecular Sequence Data , Organ Size/drug effects , Oxidative Stress/drug effects
2.
Steroids ; 77(3): 224-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22138207

ABSTRACT

Using 22R-hydroxycholesterol as a sub-structure to screen natural compound databases, we identified a naturally occurring steroid (sc-7) with a 16-acetoxy-22R-hydroxycholesterol moiety, in which the hydroxyl groups in positions 3 and 22 are esterified by an acetoxy group and in which the carbon in position 26 carries a functional diacetylamino. sc-7 is an analog of the sex steroids dehydro-oogoniol and antheridiol, can be isolated from the water mold Achlya heterosexualis, and promoted neurogenesis in vitro and in vivo. Mouse embryonic teratocarcinoma P19 cells exposed to sc-7 for 2days followed by a 5-day wash-out differentiated into cholinergic neurons that expressed specific neuronal markers and displayed axonal formation. Axons continued growing up to 28days after treatment. In vivo, infusion of sc-7 for 2weeks into the left ventricle of the rat brain followed by a 3-week wash-out induced bromodeoxyuridine uptake by cells of the ependymal layer and subventricular zone that co-localized with doublecortin and glial fibrillary acidic protein immunostaining, demonstrating induction of proliferation and differentiation of neuronal progenitors. Migrating neuroblasts were also observed in the corpus callosum. Thus, under these experimental conditions, adult ependymal cells resumed proliferation and differentiation. Taken together, these results suggest that sc-7 is an interesting molecule for stimulating in situ neurogenesis from resident neuronal progenitors as part of neuron replacement therapy. sc-7 did not bind to nuclear steroid receptors and was not metabolized as a steroid, supporting our hypothesis that the neurogenic effect of sc-7 is not likely due to a steroid-like effect.


Subject(s)
Achlya/chemistry , Cholinergic Neurons/drug effects , Neurogenesis , Steroids/chemistry , Animals , Axons/drug effects , Biomarkers/chemistry , Bromodeoxyuridine/chemistry , Cell Movement , Cell Proliferation , Cholinergic Neurons/chemistry , Corpus Callosum/chemistry , Corpus Callosum/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Ependyma/chemistry , Ependyma/drug effects , Glial Fibrillary Acidic Protein/chemistry , Hydroxycholesterols/chemistry , Immunohistochemistry , Infusions, Intraventricular , Male , Mice , Microtubule-Associated Proteins/chemistry , Neural Stem Cells/chemistry , Neural Stem Cells/drug effects , Neuropeptides/chemistry , Rats , Rats, Long-Evans , Steroids/isolation & purification , Teratocarcinoma/drug therapy , Time Factors , Tubulin/chemistry , Vimentin/chemistry
3.
Med Chem ; 6(3): 123-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20438444

ABSTRACT

We report herein the synthesis and biological evaluation of dimethyl-carbamic acid 2,3-bis-dimethylcarbamoyloxy-6-(4-ethyl-piperazine-1-carbonyl)-phenyl ester (SP-04), a new drug candidate that is designed to offer a multi-target therapeutic neuroprotective approach as a treatment for Alzheimer's disease (AD). SP-04 inhibits acetylcholinesterase (AchE) activity both in vitro and in vivo, and induces a dose-dependent increase in Ach levels. SP-04 releases the metabolite 4-(4-ethyl-piperazin-1-yl)-1-(2,3,4-trihydroxy-phenyl)-butan-1-one (SP-04m). Both SP-04 and SP-04m are s1-receptor antagonists supporting their interest in relieving symptoms related to psychosis, a non-cognitive condition often associated with AD. SP-04m displays important antioxidant properties and both SP-04 and SP-04m offers neuroprotection against Ab42 toxicity in various neuronal cell lines. In addition, both SP-04 and SP-04m protect neuronal cells and rat brain mitochondria exposed to various mitochondrial respiratory chain complex toxins. Taken together these data suggest that the SP-04 multi-targeting approach might offer a novel therapeutic strategy for the treatment of AD.


Subject(s)
Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Receptors, sigma/antagonists & inhibitors , Acetylcholinesterase , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Brain/drug effects , Carbamates/chemical synthesis , GPI-Linked Proteins/antagonists & inhibitors , Gallic Acid/analogs & derivatives , Gallic Acid/chemical synthesis , HEK293 Cells , Humans , Male , Mitochondria/drug effects , PC12 Cells , Peptide Fragments/metabolism , Piperazines/chemical synthesis , Prodrugs/chemical synthesis , Rats , Rats, Long-Evans , Sigma-1 Receptor
4.
Stem Cells Cloning ; 3: 119-27, 2010.
Article in English | MEDLINE | ID: mdl-24198517

ABSTRACT

PURPOSE: Neural stem cell transplantation as a brain repair strategy is a very promising technology. However, despite many attempts, the clinical success remains very deceiving. Despite clear evidence that sexual dimorphism rules many aspects of human biology, the occurrence of a sex difference in neural stem cell biology is largely understudied. Herein, we propose to determine whether gender is a dimension that drives the fate of neural stem cells through aging. Should it occur, we believe that neural stem cell sexual dimorphism and its variation during aging should be taken into account to refine clinical approaches of brain repair strategies. METHODS: Neural stem cells were isolated from the subventricular zone of three- and 20-month-old male and female Long-Evans rats. Expression of the estrogen receptors, ERα and ERß, progesterone receptor, androgen receptor, and glucocorticoid receptor was analyzed and quantified by Western blotting on undifferentiated neural stem cells. A second set of neural stem cells was treated with retinoic acid to trigger differentiation, and the expression of neuronal, astroglial, and oligodendroglial markers was determined using Western blotting. CONCLUSION: We provided in vitro evidence that the fate of neural stem cells is affected by sex and aging. Indeed, young male neural stem cells mainly expressed markers of neuronal and oligodendroglial fate, whereas young female neural stem cells underwent differentiation towards an astroglial phenotype. Aging resulted in a lessened capacity to express neuron and astrocyte markers. Undifferentiated neural stem cells displayed sexual dimorphism in the expression of steroid receptors, in particular ERα and ERß, and the expression level of several steroid receptors increased during aging. Such sexual dimorphism might explain, at least in part, the sex difference in neural fate we observed in young and old neural stem cells. These results suggest that sex and aging are two factors to be taken into consideration for future neural stem cell transplantation protocols in brain repair strategies.

5.
Stem Cells Cloning ; 3: 175-82, 2010.
Article in English | MEDLINE | ID: mdl-24198523

ABSTRACT

PURPOSE: Neural stem cell (NSC) transplantation and pharmacologic activation of endogenous neurogenesis are two approaches that trigger a great deal of interest as brain repair strategies. However, the success rate of clinical attempts using stem cells to restore neurologic functions altered either after traumatic brain injury or as a consequence of neurodegenerative disease remains rather disappointing. This suggests that factors affecting the fate of grafted NSCs are largely understudied and remain to be characterized. We recently reported that aging differentially affects the neurogenic properties of male and female NSCs. Although the sex steroids androgens and estrogens participate in the regulation of neurogenesis, to our knowledge, research on how gender-based differences affect the capacity of NSCs to differentiate and condition their neural fate is lacking. In the present study, we explored further the role of cell sex as a determining factor of the neural fate followed by differentiating NSCs and its relationship with a potential differential expression of aromatase (CYP19), the testosterone-metabolizing enzyme. RESULTS: Using NSCs isolated from the subventricular zone of three-month-old male and female Long-Evans rats and maintained as neurospheres, we showed that differentiation triggered by retinoic acid resulted in a neural phenotype that depends on cell sex. Differentiated male NSCs mainly expressed markers of neuronal fate, including ßIII-tubulin, microtubule associated protein 2, growth-associated protein 43, and doublecortin. In contrast, female NSCs essentially expressed the astrocyte marker glial fibrillary acidic protein. Quantification of the expression of aromatase showed a very low level of expression in undifferentiated female NSCs, whereas aromatase expression in male NSCs was 14-fold greater than the female level. CONCLUSION: Our results confirm our previous data that the neural phenotype acquired by differentiating NSCs largely depends on cell sex, and that differential expression of aromatase in undifferentiated NSCs might contribute to this sex-based dimorphism. Although still preliminary, our discovery may have clinical application in the development of future brain repair strategies.

6.
J Pathol ; 198(2): 157-62, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237874

ABSTRACT

Cyclin D1 and E2F-1 proteins are essential for the regulation of the G1/S transition through the cell cycle. Cyclin D1, a product of the bcl-1 gene, phosphorylates the retinoblastoma protein, releasing E2F-1, which in turn activates genes involved in DNA synthesis. Expression patterns of E2F-1 protein in thyroid proliferations have not been reported. This study used monoclonal antibodies for cyclin D1 and E2F-1 proteins to immunostain sections of normal thyroid, hyperplastic (cellular) nodules, follicular adenomas, follicular carcinomas, and papillary carcinomas. The proliferation rate was examined using an antibody specific for the Ki-67 antigen. Fluorescence in situ hybridization (FISH) methods and chromosome 11-specific probes were also employed to determine chromosome copy number and to assess for evidence of amplification at the 11q13 locus in papillary and follicular carcinomas with cyclin D1 overexpression. Concurrent overexpression of Ki-67, cyclin D1, and E2F-1 was found in the majority of benign and malignant thyroid lesions, compared with normal thyroid tissue. Cyclin D1 up-regulation was not due to extra copies of chromosome 11, or bcl-1 gene amplification. Malignant tumours showed the highest expression for all three markers, particularly papillary carcinomas. E2F-1 was detected at the same or slightly lower levels than cyclin D1. It was only found when cyclin D1 was overexpressed. Because cyclin D1 normally activates E2F-1, up-regulation of cyclin D1 may lead to E2F-1 overexpression in benign and malignant thyroid lesions.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Neoplasm Proteins/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/pathology , Adenoma/metabolism , Adenoma/pathology , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Division , Cyclin D1/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Humans , Hyperplasia/metabolism , In Situ Hybridization, Fluorescence , Ki-67 Antigen/metabolism , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...