Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Clin Sci (Lond) ; 121(9): 405-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21605084

ABSTRACT

We tested whether inhibition of mitochondrial membrane potential dissipation by CsA (ciclosporin A) would prevent doxorubicin-induced myocardial and mitochondrial dysfunction. Acute and subchronic models of doxorubicin exposition were performed in mice with either a single intraperitoneal bolus (10 mg/kg of body weight, intraperitoneal) or one injection of 4 mg·kg(-1) of body weight·week(-1) during 5 weeks. Follow-up was at 1.5 weeks and 16 weeks in acute and subchronic models respectively. Mice received either CsA (1 mg/kg of body weight, intraperitoneal on alternate days) or saline until follow-up. Heart function was evaluated by echocardiography. Mitochondrial measurements included oxygen consumption, membrane potential and externally added calcium-induced mitochondrial permeability transition. Mitochondrial mass was evaluated by transmission electronic microscopy and mtDNA (mitochondrial DNA) content. Mitochondrial dynamics were detected as the expression of GTPases involved in mitochondrial fusion and fission. In both the acute and chronic models, doxorubicin decreased left ventricular fractional shortening and survival. Heart function and survival were improved by CsA, but not by tacrolimus (FK506), a ciclosporin derivative with no inhibitory effect on the mitochondrial transition pore. In the acute model, doxorubicin exposure was associated with increased mtDNA content, mitochondrial fragmentation and changes in mitochondrial fusion- and fission-related transcripts [increases in Mfn2 (mitofusin 2), Opa1 (optic atrophy 1 homologue) and Fis1 (fission 1 homologue), and no changes in Drp1 (dynamin 1-like)]. CsA did not alter mitochondrial biogenesis, but prevented mitochondrial fragmentation and partially restored the mitochondrial energy-producing capacity. These findings suggest that in vivo CsA treatment may limit MPTP (mitochondrial permeability transition pore) opening, mitochondrial potential loss and contractile depression in acute and chronic models of cardiac toxicity induced by doxorubicin.


Subject(s)
Cyclosporine/pharmacology , Doxorubicin/adverse effects , Heart Diseases/chemically induced , Heart Diseases/pathology , Mitochondria, Heart/metabolism , Myocardium/pathology , Animals , Antibiotics, Antineoplastic/pharmacology , DNA Primers/genetics , DNA, Mitochondrial/metabolism , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Permeability , Reverse Transcriptase Polymerase Chain Reaction
2.
Endocrinology ; 151(4): 1760-72, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20133455

ABSTRACT

In the ever-changing physiological context of the neuroendocrine brain, the mechanisms by which cellular events involving neurons, astroglia, and vascular cells are coordinated to bring forth the appropriate neuronal signaling is not yet known but is amenable to examination. In the median eminence of the hypothalamus, endothelial cells are key players in the plasticity of tanycytes (specialized astroglia) and neuroendocrine synapse efficacy. Here we report that estradiol acts on both purified endothelial cells and isolated tanycytes to trigger endothelial-to-glial communication that leads to a sudden and massive retraction of tanycyte processes. The blockade of endothelial nitric oxide synthase by in vitro adenoviral-mediated gene transfer of a dominant-negative form of endothelial nitric oxide synthase abrogates the estradiol-induced tanycyte plasticity mediated by endothelial cells. In parallel, increases in prostaglandin-E(2) (PGE(2)) due to changes in cyclooxygenase (COX)-1 and COX-2 expression induced by the exposure of tanycytes to estradiol promote acute tanycyte plasticity. We also demonstrate by electron microscopy that the administration of PGE(2) to median eminence explants induces rapid neuroglial plasticity at the neurovascular junction of neurons that release GnRH (the neuropeptide controlling reproduction). Conversely, preventing local PGE(2) synthesis in the median eminence of adult female rats with the COX inhibitor indomethacin impairs the ovarian cycle, a process that requires a pulsatile, coordinated delivery of GnRH into the hypothalamo-hypophyseal portal system. Taken together, our findings show that estradiol controls the dialog between endothelial cells and astroglia to regulate neuroglial plasticity in the neuroendocrine brain.


Subject(s)
Cell Shape/physiology , Endothelial Cells/physiology , Ependyma/physiology , Estradiol/physiology , Median Eminence/physiology , Neuroglia/physiology , Analysis of Variance , Animals , Blotting, Western , Cell Communication/drug effects , Cell Communication/physiology , Cell Culture Techniques , Cell Shape/drug effects , Cells, Cultured , Dinoprostone/pharmacology , Endothelial Cells/drug effects , Ependyma/drug effects , Estradiol/pharmacology , Hypothalamo-Hypophyseal System/physiology , Neuroglia/drug effects , Nitric Oxide Synthase Type III/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Rats , Rats, Sprague-Dawley
3.
PLoS One ; 4(3): e4843, 2009.
Article in English | MEDLINE | ID: mdl-19290042

ABSTRACT

The role of Tau phosphorylation in neurofibrillary degeneration linked to Alzheimer's disease remains to be established. While transgenic mice based on FTDP-17 Tau mutations recapitulate hallmarks of neurofibrillary degeneration, cell models could be helpful for exploratory studies on molecular mechanisms underlying Tau pathology. Here, "human neuronal cell lines" overexpressing Wild Type or mutated Tau were established. Two-dimensional electrophoresis highlights that mutated Tau displayed a specific phosphorylation pattern, which occurs in parallel to the formation of Tau clusters as visualized by electron microscopy. In fact, this pattern is also displayed before Tau pathology onset in a well established mouse model relevant to Tau aggregation in Alzheimer's disease. This study suggests first that pathological Tau mutations may change the distribution of phosphate groups. Secondly, it is possible that this molecular event could be one of the first Tau modifications in the neurofibrillary degenerative process, as this phenomenon appears prior to Tau pathology in an in vivo model and is linked to early steps of Tau nucleation in Tau mutants cell lines. Such cell lines consist in suitable and evolving models to investigate additional factors involved in molecular pathways leading to whole Tau aggregation.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Mutation , tau Proteins/metabolism , Animals , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Phosphorylation , Protein Conformation , tau Proteins/chemistry , tau Proteins/genetics
4.
Endocrinology ; 149(2): 587-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18006627

ABSTRACT

The activation of nitric oxide (NO) signaling pathways in hypothalamic neurons plays a key role in the control of GnRH secretion that is central to reproductive function. It is unknown whether NO directly modulates the firing behavior of GnRH neurons in the preoptic region of the mature brain. Using patch-clamp recordings from GnRH neurons expressing green fluorescent protein in adult mice brain slices, we demonstrate that the NO precursor, L-arginine (Arg), or the NO donor, diethylamine/NO, induced a robust and reversible reduction in the spontaneous firing activity of GnRH neurons, including bursting activity. The effects of L-Arg were prevented by the NO synthase inhibitor N omega-nitro-L-Arg methyl ester hydrochloride. Histochemical studies revealing a close anatomical relationship between neurons producing NO and GnRH perikarya, together with the loss of the L-Arg-mediated inhibition of GnRH neuronal activity via the selective blockade of neuronal NO synthase, suggested that the primary source of local NO production in the mouse preoptic region was neuronal. Synaptic transmission uncoupling did not alter the effect of NO, suggesting that NO affects the firing pattern of GnRH neurons by acting at a postsynaptic site. We also show that the NO-mediated changes in membrane properties in the GnRH neurons require soluble guanylyl cyclase activity and may involve potassium conductance. By revealing that NO is a direct modulator of GnRH neuronal activity, our results introduce the intriguing possibility that this gaseous neurotransmitter may be used by the sexual brain to modulate burst firing patterns. It may set into phase the bursting activity of GnRH neurons at key stages of reproductive physiology.


Subject(s)
Action Potentials/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/enzymology , Nitric Oxide Synthase Type I/metabolism , Signal Transduction/physiology , Action Potentials/drug effects , Age Factors , Animals , Arginine/pharmacology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Guanylate Cyclase/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/physiology , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Patch-Clamp Techniques , Potassium/metabolism , Signal Transduction/drug effects
5.
Mol Ther ; 15(11): 1963-72, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17712334

ABSTRACT

Most adenoviral vectors (HAdvs) elaborated for gene therapy are derived from serotype 5 viruses that use clathrin-coated vesicle endocytosis for cell entry. However, it appears that adenoviral vectors are able to take advantage of lipid raft/caveolae endocytosis to infect cells. In vivo targeting of a therapeutic gene to specific cells by vector engineering has become a major focus of gene therapy research. Yet, modification of adenoviral tropism, especially fiber gene engineering, can induce deficient intracellular trafficking of the viral particle, with a shift in subcellular localization resulting in extensive exocytosis. In this study we demonstrate that uptake of a fiber-modified adenovirus using lipid raft/caveolae endocytosis leads to non-altered intracellular trafficking without endosomal retention. Moreover, activation of lipid raft structures by this vector leads to the formation of "mega-caveosomes". These results demonstrate that, by forcing adenoviruses to take advantage of a non-clathrin, non-classical endocytic pathway, it is possible to compensate for the deficiency in endosomolysis that is associated with the use of some of the fiber-modified adenoviral constructs. Moreover, it renders such vectors ideal candidates for infecting human coxsackie and adenoviruses receptor (hCAR) negative cells.


Subject(s)
Adenoviridae/metabolism , Caveolae/metabolism , Endocytosis , Adenoviridae/genetics , Animals , Biological Transport , Cell Line , Clathrin/metabolism , Cricetinae , Cricetulus , Gene Expression , Humans , Kinetics , Microscopy, Immunoelectron , Virion/metabolism , Virion/ultrastructure
6.
Psychoneuroendocrinology ; 32 Suppl 1: S46-51, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17629628

ABSTRACT

It is becoming increasingly apparent that non-neuronal cells play a critical role in generating and regulating the flow of information within the brain. Among these non-neuronal cells, astroglial cells have been shown to play important roles in the control of both synaptic transmission and neurosecretion. In addition to modulating neuronal activity, astroglial cells interact with endothelial cells throughout the central nervous system to define specific functional domains. In the hypothalamus, neurons that release gonadotropin-releasing hormone (GnRH), the neurohormone that controls both sexual development and adult reproductive function, offer an attractive model system in which to study glial-neuronal-endothelial interactions. Within the median eminence of the hypothalamus, alterations of the anatomical relationship that exists between GnRH axon terminals and ependymoglial cell processes belonging to tanycytes regulate the direct access of GnRH neurosecretory axons to the vascular wall. This cell plasticity presumably modulates the release of GnRH into the portal vasculature during the reproductive cycle. Both structural changes and GnRH secretory activity appear to be modulated, at least in part, by specific cell-cell signalling molecules secreted by astrocytes, tanycytes and endothelial cells. It is becoming increasingly clear that among the different factors that may be involved, glial cells use growth factor members of the epidermal growth factor (EGF) family, acting via receptors endowed with tyrosine kinase activity, to produce morphological changes and release neuroactive substances that directly excite nearby neurons, whereas endothelial cells of the median eminence employ nitric oxide to induce neuroglial plasticity and facilitate GnRH release.


Subject(s)
Cell Communication/physiology , Endothelial Cells/physiology , Hypothalamus/physiology , Neuroglia/physiology , Neurons/physiology , Reproduction/physiology , Animals , Cell Differentiation/physiology , Humans , Hypothalamus/cytology , Neurosecretory Systems/physiology , Sexual Maturation/physiology
7.
J Neurosci ; 27(23): 6103-14, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17553983

ABSTRACT

Considerable research has been devoted to the understanding of how nitric oxide (NO) influences brain function. Few studies, however, have addressed how its production is physiologically regulated. Here, we report that protein-protein interactions between neuronal NO synthase (nNOS) and glutamate NMDA receptors via the scaffolding protein postsynaptic density-95 (PSD-95) in the hypothalamic preoptic region of adult female rats is sensitive to cyclic estrogen fluctuation. Coimmunoprecipitation experiments were used to assess the physical association between nNOS and NMDA receptor NR2B subunit in the preoptic region of the hypothalamus. We found that nNOS strongly interacts with NR2B at the onset of the preovulatory surge at proestrus (when estrogen levels are highest) compared with basal-stage diestrous rats. Consistently, estrogen treatment of gonadectomized female rats also increases nNOS/NR2B complex formation. Moreover, endogenous fluctuations in estrogen levels during the estrous cycle coincide with changes in the physical association of nNOS to PSD-95 and the magnitude of NO release in the preoptic region. Finally, temporary and local in vivo suppression of PSD-95 synthesis by using antisense oligodeoxynucleotides leads to inhibition of nNOS activity in the preoptic region and disrupted estrous cyclicity, a process requiring coordinated activation of neurons containing gonadotropin-releasing hormone (the neuropeptide controlling reproductive function). In conclusion, our findings identify a novel steroid-mediated molecular mechanism that enables the adult mammalian brain to control NO release under physiological conditions.


Subject(s)
Estrogens/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Reproduction/physiology , Age Factors , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Estrous Cycle/metabolism , Female , Rats
8.
J Biol Chem ; 282(25): 18197-18205, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17468104

ABSTRACT

Amyloid precursor protein (APP) metabolism is central to the pathogenesis of Alzheimer disease. We showed recently that the amyloid intracellular domain (AICD), which is released by gamma-secretase cleavage of APP C-terminal fragments (CTFs), is strongly increased in cells treated with alkalizing drugs (Vingtdeux, V., Hamdane, M., Bégard, S., Loyens, A., Delacourte, A., Beauvillain, J.-C., Buée, L., Marambaud, P., and Sergeant, N. (2007) Neurobiol. Dis. 25, 686-696). Herein, we aimed to determine the cell compartment in which AICD accumulates. We show that APP-CTFs and AICD are present in multivesicular structures. Multivesicular bodies contain intraluminal vesicles (known as exosomes) when released in the extracellular space. We demonstrate that APP, APP-CTFs, and AICD are integrated and secreted within exosomes in differentiated neuroblastoma and primary neuronal culture cells. Together with recent data showing that amyloid-beta is also found in exosomes, our data show that multivesicular bodies are essential organelles for APP metabolism and that all APP metabolites can be secreted in the extracellular space.


Subject(s)
Amyloid/metabolism , Enzyme Inhibitors/pharmacology , Animals , Brain/embryology , Cell Line, Tumor , Endosomes/metabolism , Humans , Macrolides/pharmacology , Models, Biological , Neuroblastoma/metabolism , Neurons/metabolism , Organelles/metabolism , Protein Structure, Tertiary , Rats , Rats, Wistar
9.
Neurobiol Dis ; 25(3): 686-96, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17207630

ABSTRACT

The amyloid precursor protein (APP) metabolism is central to pathogenesis of Alzheimer's disease (AD). Parenchymal amyloid deposits, a neuropathological hallmark of AD, are composed of amyloid-beta peptides (Abeta). Abeta derives from the amyloid precursor protein (APP) by sequential cleavages by beta- and gamma-secretases. Gamma-secretase cleavage releases the APP intracellular domain (AICD), suggested to mediate a nuclear signaling. Physiologically, AICD is seldom detected and thus supposed to be rapidly degraded. The mechanisms responsible of its degradation remain unknown. We used a pharmacological approach and showed that several alkalizing drugs induce the accumulation of AICD in neuroblastoma SY5Y cell lines stably expressing APP constructs. Moreover, alkalizing drugs induce AICD accumulation in naive SY5Y, HEK and COS cells. This accumulation is not mediated by the proteasome or metallopeptidases and is not the result of an increased gamma-secretase activity since the gamma-secretase cleavage of Notch1 and N-Cadherin is not affected by alkalizing drug treatments. Altogether, our data demonstrate for the first time that alkalizing drugs induce the accumulation of AICD, a mechanism likely mediated by the endosome/lysosome pathway.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Hydrogen-Ion Concentration , Alkalies/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , COS Cells , Cadherins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration/drug effects , Kidney/cytology , Lysosomes/metabolism , Macrolides/pharmacology , Neuroblastoma , Protein Structure, Tertiary , Receptors, Notch/metabolism , Solubility , Transfection
10.
J Neurochem ; 99(2): 616-27, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16899066

ABSTRACT

26RFa is a novel RFamide peptide originally isolated in the amphibian brain. The 26RFa precursor has been subsequently characterized in various mammalian species but, until now, the anatomical distribution and the molecular forms of 26RFa produced in the CNS of mammals, in particular in human, are unknown. In the present study, we have investigated the localization and the biochemical characteristics of 26RFa-like immunoreactivity (LI) in two regions of the human CNS--the hypothalamus and the spinal cord. Immunohistochemical labeling using specific antibodies against human 26RFa and in situ hybridization histochemistry revealed that in the human hypothalamus 26RFa-expressing neurons are located in the paraventricular and ventromedial nuclei. In the spinal cord, 26RFa-expressing neurons were observed in the dorsal and lateral horns. Characterization of 26RFa-related peptides showed that two distinct molecular forms of 26RFa are present in the human hypothalamus and spinal cord, i.e. 26RFa and an N-terminally elongated form of 43 amino acids designated 43RFa. These data provide the first evidence that 26RFa and 43RFa are actually produced in the human CNS. The distribution of 26RF-LI suggests that 26RFa and/or 43RFa may modulate feeding, sexual behavior and transmission of nociceptive stimuli.


Subject(s)
Hypothalamus/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Spinal Cord/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence/physiology , Animals , Chromatography, High Pressure Liquid/methods , Female , Humans , Hypothalamus/anatomy & histology , Immunohistochemistry , Male , Neurons/cytology , Neuropeptides/analysis , Neuropeptides/chemistry , PC12 Cells , Paraventricular Hypothalamic Nucleus/anatomy & histology , Paraventricular Hypothalamic Nucleus/metabolism , Posterior Horn Cells/anatomy & histology , Posterior Horn Cells/metabolism , Protein Isoforms/analysis , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary/physiology , Radioimmunoassay , Rats , Spinal Cord/anatomy & histology , Ventromedial Hypothalamic Nucleus/anatomy & histology , Ventromedial Hypothalamic Nucleus/metabolism
11.
Cancer Res ; 66(6): 3177-87, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540669

ABSTRACT

Lamellarin D is a marine alkaloid with a pronounced cytotoxicity against a large panel of cancer cell lines and is a potent inhibitor of topoisomerase I. However, lamellarin D maintains a marked cytotoxicity toward cell lines resistant to the reference topoisomerase I poison camptothecin. We therefore hypothesized that topoisomerase I is not the only cellular target for the drug. Using complementary cell-based assays, we provide evidence that lamellarin D acts on cancer cell mitochondria to induce apoptosis. Lamellarin D, unlike camptothecin, induces early disruption of the inner mitochondrial transmembrane potential (Deltapsi(m)) in the P388 leukemia cell line. The functional alterations are largely prevented by cyclosporin A, an inhibitor of the mitochondrial permeability transition (MPT), but not by the inhibitor of caspases, benzyloxycarbonyl-Val-Ala-Asp(Ome)-fluoromethylketone. Deltapsi(m) disruption is associated with mitochondrial swelling and cytochrome c leakage. Using a reliable real-time flow cytometric monitoring of Deltapsi(m) and swelling of mitochondria isolated from leukemia cells, we show that lamellarin D has a direct MPT-inducing effect. Furthermore, mitochondria are required in a cell-free system to mediate lamellarin D-induced nuclear apoptosis. The direct mitochondrial effect of lamellarin D accounts for the sensitivity of topoisomerase I-mutated P388CPT5 cells resistant to camptothecin. Interestingly, a tumor-active analogue of lamellarin D, designated PM031379, also exerts a direct proapoptotic action on mitochondria, with a more pronounced activity toward mitochondria of tumor cell lines compared with nontumor cell lines. Altogether, this work reinforces the pharmacologic interest of the lamellarins and defines lamellarin D as a lead in the search for treatments against chemoresistant cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coumarins/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Mitochondria/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Camptothecin/pharmacology , Cell Line, Tumor , Cell Membrane Permeability , Cell-Free System , Cytochromes c/metabolism , Humans , Leukemia P388/drug therapy , Leukemia P388/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , NIH 3T3 Cells , Rats
12.
Peptides ; 27(5): 1110-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16517014

ABSTRACT

A number of RFamide peptides have been characterized in invertebrate species and these peptides have been found to exert a broad spectrum of biological activities. In contrast, in vertebrates, our knowledge on RFamide peptides is far more limited and only a few members of the RFamide peptide family have been identified in various vertebrate classes during the last years. The present review focuses on two novel RFamide peptides, Rana RFamide (R-RFa) and 26RFa, that have been recently isolated from the amphibian brain. R-RFa shares the C-terminal LPLRFamide motif with other RFamide peptides previously identified in mammals, birds and fish. The distribution of R-RFa in the frog brain exhibits strong similarities with those of other LPLRFamide peptides, notably in the periventricular region of the hypothalamus. There is also evidence that the physiological functions of R-RFa and other LPLRFamide peptides have been conserved from fish to mammals; in particular, all these peptides appear to be involved in the control of pituitary hormone secretion. 26RFa does not exhibit any significant structural identity with other RFamide peptides and this peptide is the only member of the family that possesses an FRFamide motif at its C-terminus. The strong conservation of the primary structure of 26RFa from amphibians to mammals suggests that this RFamide peptide is involved in important biological functions in vertebrates. As for several other RFamide peptides, 26RFa-containing neurons are present in the hypothalamus, notably in two nuclei involved in the control of feeding behavior. Indeed, 26RFa is a potent stimulator of appetite in mammals. Concurrently, recent data suggest that 26RFa exerts various neuroendocrine regulatory activities at the pituitary and adrenal level.


Subject(s)
Hypothalamus/chemistry , Neuropeptides/chemistry , Neuropeptides/physiology , Animals , Central Nervous System/chemistry , Humans , Oligopeptides/analysis , Ranidae , Receptors, Neuropeptide/analysis
13.
Chembiochem ; 6(10): 1849-56, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16196016

ABSTRACT

We have studied the mature Alzheimer-like fibers of tau by fluorescence and NMR spectroscopy. Assembly of the protein into paired helical filaments after incubation with heparin at 37 degrees C was verified by electron microscopy and size-exclusion chromatography. NMR spectroscopy on these mature fibers revealed different regions of residual mobility for tau: the N-terminal domain was found to maintain solution-like dynamics and was followed by a large domain of decreasing mobility; finally the core region was distinguished by a solid-like character. Heteronuclear-NOE data indicate that the decreasing mobility is due to both a slowing down of the rapid nanosecond movements and the introduction of slower movements that lead to exchange broadening. Fluorescence spectroscopy confirmed the presence of this rigid core, and some degree of protection from hydrogen exchange for those residues was observed. Hence, our data give a more precise picture of the dynamics of tau when it is integrated into mature filaments and should provide further understanding of the molecular processes that govern aggregation.


Subject(s)
Neurofibrillary Tangles/chemistry , tau Proteins/chemistry , Chromatography, Gel , Heparin/chemistry , Microscopy, Electron, Transmission , Neurofibrillary Tangles/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Spectrometry, Fluorescence , tau Proteins/ultrastructure
14.
J Invest Dermatol ; 125(4): 647-58, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16185262

ABSTRACT

Terminally differentiated keratinocytes are dead enucleated squams. We showed previously that the mitochondria-dependent cell death pathway might be gradually activated as differentiation progresses. In this study, we demonstrated that protoporphyrin IX, staurosporine, and rotenone induced apoptotic-like changes in the mitochondria, and early differentiation of keratinocytes without inducing apoptosis. Kinetics studies established that differentiation-related changes, including growth arrest, flattened morphology, stratification, and keratin 10 (K10) expression, were downstream of mitochondrial depolarization and proliferation, reactive oxygen species (ROS) production, and release of cytochrome c and apoptosis-inducing factor. When these changes were prevented by overexpressing Bcl-2 or pharmacologically decreasing the ROS level, K10 upregulation was inhibited, implying that the differentiated phenotype and K10 expression require apoptotic mitochondria, ROS being the most likely differentiation-mediating factor. Our data also suggest that the same mitochondria-affecting stimuli can induce either differentiation or apoptosis, depending on the keratinocyte's competency to undergo differentiation, a competency that may be controlled by Bcl-2.


Subject(s)
Antioxidants/pharmacology , Apoptosis , Cell Differentiation , Keratinocytes/cytology , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Calcium/physiology , Cells, Cultured , Cytochromes c/metabolism , Female , Humans , Mitochondria/physiology , Protoporphyrins/pharmacology , Reactive Oxygen Species , Rotenone/pharmacology , Staurosporine/pharmacology
15.
J Neurosci ; 24(46): 10353-63, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548649

ABSTRACT

Glial and endothelial cells interact throughout the brain to define specific functional domains. Whether endothelial cells convey signals to glia in the mature brain is unknown but is amenable to examination in circumventricular organs. Here we report that purified endothelial cells of one of these organs, the median eminence of the hypothalamus, induce acute actin cytoskeleton remodeling in isolated ependymoglial cells and show that this plasticity is mediated by nitric oxide (NO), a diffusible factor. We found that both soluble guanylyl cyclase and cyclooxygenase products are involved in this endothelial-mediated control of ependymoglia cytoarchitecture. We also demonstrate by electron microscopy that activation of endogenous NO release in the median eminence induces rapid structural changes, allowing a direct access of neurosecretory axons containing gonadotropin-releasing hormone (GnRH) (the neuropeptide controlling reproductive function) to the portal vasculature. Local in vivo inhibition of NO synthesis disrupts reproductive cyclicity, a process that requires a pulsatile, coordinated delivery of GnRH into the hypothalamic-adenohypophyseal portal system. Our results identify a previously unknown function for endothelial cells in inducing neuroglial plasticity and raise the intriguing possibility that endothelial cells throughout the brain may use a similar signaling mechanism to regulate glial-neuronal interactions.


Subject(s)
Astrocytes/physiology , Brain/cytology , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Ependyma/physiology , Neuronal Plasticity/physiology , Neurosecretory Systems/cytology , Actins/ultrastructure , Animals , Astrocytes/ultrastructure , Brain/ultrastructure , Cells, Cultured , Cyclooxygenase 1 , Cyclooxygenase 2 , Cytoskeleton/ultrastructure , Endothelium, Vascular/cytology , Endothelium, Vascular/ultrastructure , Ependyma/ultrastructure , Estrous Cycle/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Guanylate Cyclase/metabolism , Median Eminence/blood supply , Median Eminence/physiology , Median Eminence/ultrastructure , Membrane Proteins , Neurosecretory Systems/ultrastructure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
J Neurochem ; 91(1): 110-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15379892

ABSTRACT

The human urotensin II (UII) precursor encompasses several potential cleavage sites and thus, processing of pro-UII may generate various forms of mature UII including the peptides of 11 (UII11), 16 (UII16) and 19 (UII19) residues. Until now, the native form of human UII had not been characterized. Here, we show that the major UII peptide occurring in the human spinal cord corresponds to UII11. In contrast, neither the UII16 nor the UII19 forms could be detected. In 50% of the brainstem and in all the spinal cord extracts analysed, a second minor UII-immunoreactive peptide was resolved. Immunohistochemical labelling of the cervical segment of the human spinal cord revealed that the UII-immunoreactive material was confined to a subset of ventral horn motoneurones. These data provide the first evidence that in the human, the UII precursor, expressed in motoneurones, is processed at the tribasic KKR93 cleavage site to generate a mature form of UII of 11 amino acids. The absence of N-terminally elongated forms of UII of 16 and 19 residues indicates that pro-UII is not cleaved at the R85 or K88 monobasic sites. Finally, the minor UII-immunoreactive peptide detected in several tissue extracts might correspond to an extended form of UII resulting from the processing of the UII precursor at the basic RK50 or RK66 doublets.


Subject(s)
Brain Stem/metabolism , Spinal Cord/metabolism , Urotensins/metabolism , Aged , Aged, 80 and over , Animals , Brain Chemistry , Chromatography, High Pressure Liquid/methods , Female , Humans , Immunohistochemistry/methods , Intracellular Signaling Peptides and Proteins , Male , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Hormones/immunology , Peptide Hormones/metabolism , Postmortem Changes , Radioimmunoassay/methods , Urotensins/analysis , Urotensins/immunology
17.
Oncogene ; 23(42): 7018-30, 2004 Sep 16.
Article in English | MEDLINE | ID: mdl-15273722

ABSTRACT

Doxorubicin is one of the most largely prescribed antitumor drug for the treatment of breast, liver and colon cancers as well as leukemia, but the cardiotoxicity of this anthracycline derivative limits its clinical use. Although doxorubicin is toxic to both cancer and cardiac cells, there are evidences suggesting that the mechanism of cell death is different for the two cell types. To investigate further this issue, we have compared the proapoptotic effects of doxorubicin and the functionally related anthracenedione compound mitoxantrone, which is also used in the clinic for the treatment of cancer. After evaluating the toxicity of the two drugs to mammary adenocarcinoma MTLn3 cells and H9C2 cardiomyocytes, we dissected the drug-induced apoptotic machinery by measuring the effects on the cell cycle progression, DNA condensation and fragmentation, production of endogenous peroxides and caspase activation. Both doxorubicin and mitoxantrone are potent inducers of apoptosis in H9C2 cardiomyocytes and MTLn3 breast cancer cells, but there are significant differences between the two cell types in terms of kinetics and order of the events. In particular, flow cytometry measurements of drug-induced changes in mitochondrial transmembrane potential and mitochondrial mass with different fluorescent probes suggested that the two drugs induced a progressive increase in mitochondrial mass in the cancer cells but not in the cardiac cells. The hypothesis was validated by means of electron microscopy, which revealed a significant increase in the number of mitochondria in drug-treated MTLn3 but not in H9C2 cells. The mitochondrial proliferation precedes the nuclear apoptosis in doxorubicin-treated MTLn3 cells. The changes in the architecture and number of mitochondria are linked to the drug-induced perturbation of the cell cycle progression and apoptosis. The proliferation of mitochondria could explain the higher toxicity of doxorubicin to cancer cells compared to cardiac cells and this suggests novel therapeutic opportunities to better control the cardiotoxicity of anthracyclines.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Doxorubicin/pharmacology , Mitochondria, Heart/ultrastructure , Mitochondria/pathology , Mitochondria/ultrastructure , Adenocarcinoma , Apoptosis/drug effects , Breast Neoplasms , Cell Line , Cell Line, Tumor , Doxorubicin/toxicity , Female , Heart/drug effects , Humans , Mitochondria/drug effects , Mitochondria, Heart/drug effects , Mitoxantrone/pharmacology
18.
J Soc Biol ; 198(1): 68-72, 2004.
Article in French | MEDLINE | ID: mdl-15146958

ABSTRACT

During the last decade, compelling evidence has been provided that, in addition of being regulated by transsynaptic inputs, GnRH neuroendocrine secretion is modulated by factors released both by glial cells and the endothelium of pituitary portal blood vessels. Glial cells exert their regulatory influence on GnRH release through the secretion of growth factors, such as TGFbetas and peptides member of the EGF family, that act either directly on GnRH neurons or require prostaglandin release from astrocytes, respectively. On the other hand vascular endothelial cells stimulate GnRH release via NO secretion. In addition, recent studies suggest that both glial cells and endothelial cells of the median eminence can modulate the direct access of GnRH neuroendocrine terminals to the vascular wall and thus control GnRH release efficiency. During the reproductive cycle, direct neurovascular contacts of GnRH nerve endings, that are engulfed in tanycytic endfeet, only occur at periods when massive GnRH release is required, i.e., at the onset of the preovulatory GnRH/LH surge on the day of proestrus. Recent in vitro and in vivo data demonstrate that both glial (TGFalpha and TGFbeta) and endothelial (NO) factors can induce such morphological plasticity. Neuro-glio-endothelial interactions at the median eminence of the hypothalamus thus appear to be key regulatory mechanisms for GnRH neuroendocrine secretion.


Subject(s)
Endothelial Cells/metabolism , Gonadotropin-Releasing Hormone/metabolism , Growth Substances/metabolism , Hypothalamus/cytology , Neuroglia/metabolism , Nitric Oxide/metabolism , Animals , Endothelium, Vascular/cytology , Epidermal Growth Factor/metabolism , Hypothalamus/metabolism , Median Eminence/cytology , Median Eminence/physiology , Pituitary Gland/blood supply , Proestrus/physiology , Transforming Growth Factor alpha/metabolism , Transforming Growth Factor beta/metabolism
19.
Endocrinology ; 145(4): 1794-801, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14670985

ABSTRACT

In vitro studies using immortalized GT1 cells suggest that hypothalamic astrocytes employ TGFbeta(1) to directly regulate the secretion of GnRH, the neurohormone that controls sexual maturation and adult reproductive function. However, whether such astrocyte-GnRH neuron signaling occurs in vivo is not clear. In the present study, we used in situ hybridization and immunohistochemistry to determine whether astrocytes and GnRH neurons express the molecular components necessary to set in motion communication processes involving TGFbeta(1) signaling. Double-labeling experiments showed that astrocytes in the male rat preoptic region (POA) expressed TGFbeta(1) mRNA and that GnRH perikarya were often found in close association with TGFbeta(1) mRNA-expressing cells. In addition, GnRH neuronal cell bodies in the POA expressed both type II TGFbeta receptors (TGFbeta-RII), which selectively bind TGFbeta, and Smad2/3, one of the primary transducers of TGFbeta signaling, suggesting that they are fully capable of responding directly to TGFbeta(1) stimulation. Consistent with this hypothesis, incubation of POA explants with TGFbeta(1) caused a significant, dose-dependent decrease in GnRH mRNA expression in individual neurons. This effect was observed within 1 h after TGFbeta(1)-treatment and was inhibited by addition of the soluble form of TGFbeta-RII to the incubation medium. In contrast, whereas both TGFbeta(1) and TGFbeta-RII mRNAs were abundantly expressed in both glial cells and capillaries in the median eminence, the projection field of GnRH neurons, TGFbeta-RII immunoreactivity was mainly restricted to the processes of tanycytes and did not colocalize with GnRH-immunoreactive fibers. This observation supports previous in vivo studies showing that TGFbeta(1) is unable to directly modulate decapeptide release from GnRH nerve terminals. Thus, astrocyte-derived TGFbeta(1) may directly influence GnRH expression and/or secretion in vivo by acting on the perikarya, but not the terminals, of GnRH neurons.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Median Eminence/metabolism , Transforming Growth Factor beta/physiology , Animals , Astrocytes/metabolism , Gene Expression , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/cytology , Hypothalamus, Middle/metabolism , Male , Neurons/metabolism , Phenotype , Preoptic Area/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
20.
Proc Natl Acad Sci U S A ; 100(25): 15247-52, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14657341

ABSTRACT

A neuropeptide was isolated from a frog brain extract by HPLC purification and characterized by mass spectrometry. This 26-aa neuropeptide, which belongs to the RFamide peptide family, was designated 26RFa, and its primary structure was established as VGTALGSLAEELNGYNRKKGGFSFRF-NH2. Research in databases revealed the presence of sequences homologous to frog 26RFa in the human genome and in rat ESTs. On the basis of this sequence information, the cDNAs encoding the human and rat 26RFa precursors were cloned. The two preproteins show a similar organization, with the 26RFa sequence located in the C-terminal region of the precursor. Human preprotein (prepro)-26RFa encodes an additional putative RFamide peptide that is not found in the rat precursor. The primary structures of human, rat, and frog 26RFa exhibit approximately 80% identity, and the C-terminal octapeptide has been fully conserved from amphibians to mammals. In situ hybridization histochemistry revealed that, in the rat brain, the 26RFa gene is exclusively expressed in the ventromedial hypothalamic nucleus and in the lateral hypothalamic area. 26RFa induced a dose-dependent stimulation in cAMP production by rat pituitary cells in vitro and markedly increased food intake in mice. The conservation of the primary structure of 26RFa during vertebrate evolution, the discrete localization of the mRNA encoding its precursor in hypothalamic nuclei involved in the control of feeding behavior, and the observation that 26RFa possesses orexigenic properties indicate that this neuropeptide may play important biological functions.


Subject(s)
Nerve Tissue Proteins/chemistry , Neuropeptides/chemistry , Peptides/chemistry , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Chromatography, High Pressure Liquid , Cloning, Molecular , Cyclic AMP/metabolism , DNA, Complementary/metabolism , Databases as Topic , Dose-Response Relationship, Drug , Expressed Sequence Tags , Genome, Human , Humans , Hypothalamus/metabolism , In Situ Hybridization , Male , Mass Spectrometry , Mice , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Peptide Biosynthesis , RNA, Messenger/metabolism , Ranidae , Rats , Rats, Wistar , Sequence Homology, Amino Acid , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...