Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 159(7): 1475-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20136839

ABSTRACT

BACKGROUND AND PURPOSE: Neuronal ion channels are key targets of general anaesthetics and alcohol, and binding of these drugs to pre-existing and relatively specific sites is thought to alter channel gating. However, the underlying molecular mechanisms of this action are still poorly understood. Here, we investigated the neuronal Shaw2 voltage-gated K(+) (K(v)) channel to ask whether the inhalational anaesthetic halothane and n-alcohols share a binding site near the activation gate of the channel. EXPERIMENTAL APPROACH: Focusing on activation gate mutations that affect channel modulation by n-alcohols, we investigated n-alcohol-sensitive and n-alcohol-resistant K(v) channels heterologously expressed in Xenopus oocytes to probe the functional modulation by externally applied halothane using two-electrode voltage clamping and a gas-tight perfusion system. KEY RESULTS: Shaw2 K(v) channels are reversibly inhibited by halothane in a dose-dependent and saturable manner (K(0.5)= 400 microM; n(H)= 1.2). Also, discrete mutations in the channel's S4S5 linker are sufficient to reduce or confer inhibition by halothane (Shaw2-T330L and K(v)3.4-G371I/T378A respectively). Furthermore, a point mutation in the S6 segment of Shaw2 (P410A) converted the halothane-induced inhibition into halothane-induced potentiation. Lastly, the inhibition resulting from the co-application of n-butanol and halothane is consistent with the presence of overlapping binding sites for these drugs and weak binding cooperativity. CONCLUSIONS AND IMPLICATIONS: These observations strongly support a molecular model of a general anaesthetic binding site in the Shaw2 K(v) channel. This site may involve the amphiphilic interface between the S4S5 linker and the S6 segment, which plays a pivotal role in K(v) channel activation.


Subject(s)
Alcohols/pharmacology , Anesthetics, Inhalation/pharmacology , Halothane/pharmacology , Neurons/drug effects , Shaw Potassium Channels/drug effects , Animals , Mutation , Neurons/metabolism , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Xenopus laevis
2.
Proc Natl Acad Sci U S A ; 105(46): 18029-34, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-19004782

ABSTRACT

Two related neurodegenerative disorders, Western Pacific amyotrophic lateral sclerosis (ALS) and parkinsonism-dementia (PD), originally occurred at a high incidence on Guam, in the Kii peninsula of Japan, and in southern West New Guinea more than 50 years ago. These three foci shared a unique mineral environment characterized by the presence of severely low levels of Ca(2+) and Mg(2+), coupled with high levels of bioavailable transition metals in the soil and drinking water. Epidemiological studies suggest that genetic factors also contribute to the etiology of these disorders. Here, we report that a variant of the transient receptor potential melastatin 2 (TRPM2) gene may confer susceptibility to these diseases. TRPM2 encodes a calcium-permeable cation channel highly expressed in the brain that has been implicated in mediating cell death induced by oxidants. We found a heterozygous variant of TRPM2 in a subset of Guamanian ALS (ALS-G) and PD (PD-G) cases. This variant, TRPM2(P1018L), produces a missense change in the channel protein whereby proline 1018 (Pro(1018)) is replaced by leucine (Leu(1018)). Functional studies revealed that, unlike WT TRPM2, P1018L channels inactivate. Our results suggest that the ability of TRPM2 to maintain sustained ion influx is a physiologically important function and that its disruption may, under certain conditions, contribute to disease states.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Mutant Proteins/metabolism , Parkinsonian Disorders/metabolism , TRPM Cation Channels/metabolism , Adenosine Diphosphate Ribose/pharmacology , Amino Acid Sequence , Amino Acids, Diamino/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Conserved Sequence , Cyanobacteria Toxins , Evolution, Molecular , Guam , Humans , Hydrogen Peroxide/pharmacology , Ion Channel Gating/drug effects , Leucine/genetics , Magnesium/pharmacology , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Oxidative Stress/drug effects , Proline/genetics , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics , Temperature
3.
Neurosci Lett ; 436(2): 171-6, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18395978

ABSTRACT

PRA1 domain family, member 2 (PRAF2) is a novel 19-kDa protein with a prenylated Rab acceptor 1 (PRA1) motif and four transmembrane domains. Our previous studies revealed that PRAF2 is highly expressed in the brain and serves as a candidate prognostic marker in neuroblastoma (NB). PRAF2 is related to proteins PRAF1 (PRA1, prenylin, Yip3) and PRAF3 (GTRAP3-18, JWA, Arl6-IP5), both of which are enriched in the brain and implicated in cellular transport and endo/exocytic vesicle trafficking. However, the function for PRAF2 remains unknown. In this study, we analyzed the distribution and localization of PRAF2 in the mature human brain using two new antibodies specific for the protein. Analysis by immunohistochemistry revealed that in the human cerebellum, the PRAF2 protein was strongly expressed in Purkinje cells and, more moderately, in cells of the molecular and the granular layers. In the cerebral cortex, hippocampus, and lateral ventricles, PRAF2 protein was detected in neuronal cells, but not in non-neuronal cells. Intriguingly, immunoblot analysis revealed that PRAF2 is enriched in synaptic vesicles (SVs) prepared from rat brains. The expression of PRAF2 in specific regions of the brain including SVs suggest an important physiological function for this novel protein, possibly by participating in multiple aspects of SV maturation, transport, and signal transmission.


Subject(s)
Brain/metabolism , Brain/ultrastructure , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Synaptic Vesicles/metabolism , Brain/anatomy & histology , Cell Line, Tumor , Gene Expression/physiology , Humans , Neuroblastoma
4.
Clin Cancer Res ; 13(21): 6312-9, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17975142

ABSTRACT

PURPOSE: Prenylated Rab acceptor 1 domain family, member 2 (PRAF2) is a novel 19-kDa protein that has recently been implicated in human cancer. In the present study, we analyzed for the first time PRAF2 mRNA expression in a large set of human tumors. The high expression in neuroblastic tumors prompted us to analyze PRAF2 expression correlations with genetic and clinical features of these tumors. In addition, we determined the localization of PRAF2 protein in neuroblastoma cells and studied its regulation in apoptosis. EXPERIMENTAL DESIGN: Affymetrix microarray analysis was done with a set of 41 different tumor types (1,426 samples) in the public domain, a set of three different neuroblastic tumor types (110 samples), and a panel of 25 neuroblastoma cell lines. The subcellular localization of endogenous PRAF2 in neuroblastoma cells was identified by immunofluorescence microscopy and apoptosis detected by Annexin V staining and poly(ADP-ribose) polymerase cleavage. RESULTS: PRAF2 mRNA was detected in 970 of 1,426 samples in the public data set. All 110 neuroblastic tumors expressed PRAF2 at higher levels than any other tumor examined. Importantly, PRAF2 expression levels significantly correlated with the following clinical features: patient age at diagnosis (P = 6.19 x 10(-5)), survival (P = 1.32 x 10(-3)), International Neuroblastoma Staging System stage (P = 2.86 x 10(-4)), and MYCN amplification (P = 3.74 x 10(-3)). PRAF2 localized in bright cytoplasmic punctae and protein levels increased in neuroblastoma cells that underwent cerulenin-induced apoptosis. CONCLUSIONS: Elevated PRAF2 expression levels correlated with unfavorable genetic and clinical features, suggesting PRAF2 as a candidate prognostic marker of neuroblastoma.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Cerulenin/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neuroblastoma/metabolism , Cell Line, Tumor , Child, Preschool , Endosomes/metabolism , Humans , Infant , Infant, Newborn , Microscopy, Fluorescence , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...