Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Am J Physiol Endocrinol Metab ; 319(4): E678-E688, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32776828

ABSTRACT

Testosterone supplementation during energy deficit promotes whole body lean mass accretion, but the mechanisms underlying that effect remain unclear. To elucidate those mechanisms, skeletal muscle molecular adaptations were assessed from muscle biopsies collected before, 1 h, and 6 h after exercise and a mixed meal (40 g protein, 1 h postexercise) following 14 days of weight maintenance (WM) and 28 days of an exercise- and diet-induced 55% energy deficit (ED) in 50 physically active nonobese men treated with 200 mg testosterone enanthate/wk (TEST) or placebo (PLA) during the ED. Participants (n = 10/group) exhibiting substantial increases in leg lean mass and total testosterone (TEST) were compared with those exhibiting decreases in both of these measures (PLA). Resting androgen receptor (AR) protein content was higher and fibroblast growth factor-inducible 14 (Fn14), IL-6 receptor (IL-6R), and muscle ring-finger protein-1 gene expression was lower in TEST vs. PLA during ED relative to WM (P < 0.05). Changes in inflammatory, myogenic, and proteolytic gene expression did not differ between groups after exercise and recovery feeding. Mechanistic target of rapamycin signaling (i.e., translational efficiency) was also similar between groups at rest and after exercise and the mixed meal. Muscle total RNA content (i.e., translational capacity) increased more during ED in TEST than PLA (P < 0.05). These findings indicate that attenuated proteolysis at rest, possibly downstream of AR, Fn14, and IL-6R signaling, and increased translational capacity, not efficiency, may drive lean mass accretion with testosterone administration during energy deficit.


Subject(s)
Energy Metabolism/drug effects , Protein Modification, Translational/drug effects , Receptors, Androgen/biosynthesis , Testosterone/pharmacology , Adolescent , Adult , Body Composition , Diet , Exercise , Hormones/blood , Humans , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Receptors, Interleukin-6/metabolism , TWEAK Receptor/metabolism , Up-Regulation , Young Adult
2.
Fungal Genet Biol ; 134: 103281, 2020 01.
Article in English | MEDLINE | ID: mdl-31626987

ABSTRACT

Cadmium (Cd) is a heavy metal present in the environment mainly as a result of industrial contamination that can cause toxic effects to life. Some microorganisms, as Trichoderma harzianum, a fungus used in biocontrol, are able to survive in polluted environments and act as bioremediators. Aspects about the tolerance to the metal have been widely studied in other fungi although there are a few reports about the response of T. harzianum. In this study, we determined the effects of cadmium over growth of T. harzianum and used RNA-Seq to identify significant genes and processes regulated in the metal presence. Cadmium inhibited the fungus growth proportionally to its concentration although the fungus exhibited tolerance as it continued to grow, even in the highest concentrations used. A total of 3767 (1993 up and 1774 down) and 2986 (1606 up and 1380 down) differentially expressed genes were detected in the mycelium of T. harzianum cultivated in the presence of 1.0 mg mL-1 or 2.0 mg mL-1 of CdCl2, respectively, compared to the absence of the metal. Of these, 2562 were common to both treatments. Biological processes related to cellular homeostasis, transcription initiation, sulfur compound biosynthetic and metabolic processes, RNA processing, protein modification and vesicle-mediated transport were up-regulated. Carbohydrate metabolic processes were down-regulated. Pathway enrichment analysis indicated induction of glutathione and its precursor's metabolism. Interestingly, it also indicated an intense transcriptional induction, especially by up-regulation of spliceosome components. Carbohydrate metabolism was repressed, especially the mycoparasitism-related genes, suggesting that the mycoparasitic ability of T. harzianum could be affected during cadmium exposure. These results contribute to the advance of the current knowledge about the response of T. harzianum to cadmium exposure and provide significant targets for biotechnological improvement of this fungus as a bioremediator and a biocontrol agent.


Subject(s)
Cadmium/pharmacology , Gene Expression Regulation, Fungal/drug effects , Genes, Fungal , Hypocreales/drug effects , Hypocreales/genetics , Transcriptome/drug effects , Carbohydrate Metabolism/genetics , Hypocreales/growth & development , Mycelium/drug effects , Mycelium/genetics , Mycelium/growth & development , Protein Modification, Translational/drug effects , RNA Processing, Post-Transcriptional/drug effects , Spliceosomes/drug effects
3.
PLoS Comput Biol ; 15(7): e1007225, 2019 07.
Article in English | MEDLINE | ID: mdl-31323035

ABSTRACT

Exposure to the environmental toxin ß-methylamino-L-alanine (BMAA) is linked to amyotrophic lateral sclerosis (ALS), but its disease-promoting mechanism remains unknown. We propose that incorporation of BMAA into the ALS-linked protein Cu,Zn superoxide dismutase (SOD1) upon translation promotes protein misfolding and aggregation, which has been linked to ALS onset and progression. Using molecular simulation and predictive energetic computation, we demonstrate that substituting any serine with BMAA in SOD1 results in structural destabilization and aberrant dynamics, promoting neurotoxic SOD1 aggregation. We propose that translational incorporation of BMAA into SOD1 is directly responsible for its toxicity in neurodegeneration, and BMAA modification of SOD1 may serve as a biomarker of ALS.


Subject(s)
Amino Acids, Diamino/pharmacokinetics , Amino Acids, Diamino/toxicity , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/metabolism , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/genetics , Binding Sites/genetics , Computational Biology , Cyanobacteria Toxins , Enzyme Stability/genetics , Humans , Molecular Dynamics Simulation , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Folding/drug effects , Protein Modification, Translational/drug effects , Protein Modification, Translational/genetics , Protein Structure, Quaternary , Superoxide Dismutase-1/genetics
4.
J Neurochem ; 147(6): 831-848, 2018 12.
Article in English | MEDLINE | ID: mdl-30152072

ABSTRACT

For more than 150 years, it is known that occupational overexposure of manganese (Mn) causes movement disorders resembling Parkinson's disease (PD) and PD-like syndromes. However, the mechanisms of Mn toxicity are still poorly understood. Here, we demonstrate that Mn dose- and time-dependently blocks the protein translation of amyloid precursor protein (APP) and heavy-chain Ferritin (H-Ferritin), both iron homeostatic proteins with neuroprotective features. APP and H-Ferritin are post-transcriptionally regulated by iron responsive proteins, which bind to homologous iron responsive elements (IREs) located in the 5'-untranslated regions (5'-UTRs) within their mRNA transcripts. Using reporter assays, we demonstrate that Mn exposure repressed the 5'-UTR-activity of APP and H-Ferritin, presumably via increased iron responsive proteins-iron responsive elements binding, ultimately blocking their protein translation. Using two specific Fe2+ -specific probes (RhoNox-1 and IP-1) and ion chromatography inductively coupled plasma mass spectrometry (IC-ICP-MS), we show that loss of the protective axis of APP and H-Ferritin resulted in unchecked accumulation of redox-active ferrous iron (Fe2+ ) fueling neurotoxic oxidative stress. Enforced APP expression partially attenuated Mn-induced generation of cellular and lipid reactive oxygen species and neurotoxicity. Lastly, we could validate the Mn-mediated suppression of APP and H-Ferritin in two rodent in vivo models (C57BL6/N mice and RjHan:SD rats) mimicking acute and chronic Mn exposure. Together, these results suggest that Mn-induced neurotoxicity is partly attributable to the translational inhibition of APP and H-Ferritin resulting in impaired iron metabolism and exacerbated neurotoxic oxidative stress. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Amyloid beta-Protein Precursor/antagonists & inhibitors , Apoferritins/antagonists & inhibitors , Iron/metabolism , Manganese Poisoning/metabolism , 5' Untranslated Regions , Amyloid beta-Protein Precursor/metabolism , Animals , Apoferritins/metabolism , Cell Line , Cell Survival/drug effects , Humans , Mice , Mice, Inbred C57BL , Oxidative Stress , Protein Modification, Translational/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
5.
Sci Rep ; 7: 45287, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28349922

ABSTRACT

S009-131, a coumarin-chalcone hybrid, had been shown to possess anti-proliferative and anti-tumour effect by triggering apoptosis. In this report, we investigated role of DNA damage signalling pathway in S009-131 induced cancer cell death. Here we show that S009-131 causes DNA damage by potential binding to the minor groove which led to the phosphorylation and activation of ATM and DNA-PK, but not ATR, at earlier time points in order to initiate repair process. S009-131 induced DNA damage response triggered activation of p53 through phosphorylation at its key residues. Pharmacological inhibition of PIKKs abrogated S009-131 induced phosphorylation of p53 at Ser 15. DNA damage induced phosphorylation resulted in reduced proteasomal degradation of p53 by disrupting p53-MDM2 interaction. Additionally, our docking studies revealed that S009-131 might also contribute to increased cellular p53 level by occupying p53 binding pocket of MDM2. Posttranslational modifications of p53 upon S009-131 treatment led to enhanced affinity of p53 towards responsive elements (p53-RE) in the promoter regions of target genes and increased transcriptional efficiency. Together, the results suggest that S009-131 cleaves DNA through minor groove binding and eventually activates PIKKs associated DNA damage response signalling to promote stabilization and enhanced transcriptional activity of p53 through posttranslational modifications at key residues.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Coumarins/pharmacology , DNA Damage/drug effects , DNA/chemistry , Protein Modification, Translational/drug effects , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Binding Sites/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , HCT116 Cells , Histones/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitination/drug effects
6.
Bioorg Med Chem Lett ; 25(7): 1464-70, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25748161

ABSTRACT

Autophagy is defined as an evolutionarily conserved process responsible for degradation of the cytoplasmic components including protein aggregates via the lysosomal machinery. Increasing evidence has linked defective autophagic degradation of protein aggregates with the pathogenesis of neurodegenerative disorders, and it is suggested that promotion of autophagy is regarded as a potential therapeutic for these diseases including Parkinson's disease (PD). Here we identified, 3-anhydro-6-hydroxy-ophiobolin A (X15-2), an ophiobolin derivative from Bipolaris oryzae that can strongly induce autophagic degradation of α-synuclein, the major constituent of Lewy bodies. We showed that X15-2 induced autophagy is dependent on both Beclin1 and Beclin2. Knockout of ATG5 by CRISPER/Cas9 prevented X15-2 induced autophagy and degradation of α-synuclein. Mechanistically, we showed that X15-2 induces ROS and the activation of JNK signaling for the autophagic degradation of α-synuclein in PC12 cells.


Subject(s)
Ascomycota/chemistry , Autophagy/drug effects , Protein Modification, Translational/drug effects , Proteolysis/drug effects , Sesterterpenes/pharmacology , alpha-Synuclein/metabolism , Animals , Dose-Response Relationship, Drug , HeLa Cells , Humans , Molecular Conformation , PC12 Cells , Rats , Sesterterpenes/chemistry , Sesterterpenes/isolation & purification , Structure-Activity Relationship
7.
Biochem Pharmacol ; 88(1): 46-57, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24412276

ABSTRACT

Processing mutations that inhibit folding and trafficking of CFTR are the main cause of cystic fibrosis. Repair of CFTR mutants requires an understanding of the mechanisms of misfolding caused by processing mutations. Previous studies on helix-loop-helix fragments of the V232D processing mutation suggested that its mechanism was to lock transmembrane (TM) segments 3 and 4 together by a non-native hydrogen bond (Asp232(TM4)/Gln207(TM3)). Here, we performed mutational analysis to test for Asp232/Gln207 interactions in full-length CFTR. The rationale was that a V232N mutation should mimic V232D and a V232D/Q207A mutant should mature if the processing defect was caused by hydrogen bonds. We report that only Val232 mutations to charged amino acids severely blocked CFTR maturation. The V232N mutation did not mimic V232D as V232N showed 40% maturation compared to 2% for V232D. Mutation of Val232 to large nonpolar residues (Leu, Phe) had little effect. The Q207L mutation did not rescue V232D because Q207L showed about 50% maturation in the presence of corrector VX-809 while V232D/Q207A could no longer be rescued. These results suggest that V232D inhibits maturation by disrupting a hydrophobic pocket between TM segments rather than forming a non-native hydrogen bond. Disulfide cross-linking analysis of cysteines W356C(TM6) and W1145C(TM12) suggest that the V232D mutation inhibits maturation by trapping CFTR as a partially folded intermediate. Since correctors can efficiently rescue V232D CFTR, the results suggest that hydrophilic processing mutations facing a hydrophobic pocket are good candidates for rescue with pharmacological chaperones.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/genetics , Mutation , Protein Folding , Protein Modification, Translational/genetics , Amino Acid Substitution , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , HEK293 Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mutation/drug effects , Protein Folding/drug effects , Protein Modification, Translational/drug effects , Protein Structure, Secondary
8.
Osteoarthritis Cartilage ; 22(2): 259-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24333294

ABSTRACT

OBJECTIVE: There is increasing evidence that the addition of O-linked N-acetylglucosamine (O-GlcNAc) to proteins plays an important role in cell signaling pathways. In chondrocytes, accumulation of O-GlcNAc-modified proteins induces hypertrophic differentiation. Osteoarthritis (OA) is characterized by cartilage degradation, and hypertrophic-like changes in hyaline chondrocytes. However, the mechanisms responsible for these changes have not been described. Our aim was to study whether O-GlcNAcylation and the enzymes responsible for this modification are dysregulated in the cartilage of patients with knee OA and whether interleukin-1 could induce these modifications in cultured human OA chondrocytes (HOC). DESIGN: Human cartilage was obtained from patients with knee OA and from age and sex-matched healthy donors. HOC were cultured and stimulated with the catabolic cytokine IL-1α. Global protein O-GlcNAcylation and the synthesis of the key enzymes responsible for this modification, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), were assessed by western blot. RESULTS: OA was associated with a 4-fold increase in the global O-GlcNAcylation in the cartilage. OA cartilage showed a re-distribution of the OGT and OGA isoforms, with a net increase in the presence of both enzymes, in comparison to healthy cartilage. In HOC, IL-1α stimulation rapidly increased O-GlcNAcylation and OGT and OGA synthesis. CONCLUSIONS: Our results indicate that a proinflammatory milieu could favor the accumulation of O-GlcNAcylated proteins in OA cartilage, together with the dysregulation of the enzymes responsible for this modification. The increase in O-GlcNAcylation could be responsible, at least partially, for the re-expression of hypertrophic differentiation markers that have been observed in OA.


Subject(s)
Acetylglucosamine/metabolism , Cartilage, Articular/metabolism , Osteoarthritis, Knee/metabolism , Protein Modification, Translational/physiology , Acylation , Adult , Cartilage, Articular/pathology , Case-Control Studies , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Female , Humans , Inflammation Mediators/pharmacology , Interleukin-1/pharmacology , Isoenzymes/biosynthesis , Male , Middle Aged , N-Acetylglucosaminyltransferases/biosynthesis , Osteoarthritis, Knee/pathology , Protein Modification, Translational/drug effects , beta-N-Acetylhexosaminidases/biosynthesis
9.
Proteomics ; 13(5): 833-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23303650

ABSTRACT

Viroids are single-stranded, circular, noncoding RNAs that infect plants, causing devastating diseases. In this work, we employed 2D DIGE, followed by MS identification, to analyze the response of tomato plants infected by Citrus exocortis viroid (CEVd). Among the differentially expressed proteins detected, 45 were successfully identified and classified into different functional categories. Validation results by RT-PCR allowed us to classify the proteins into two expression groups. First group included genes with changes at the transcriptional level upon CEVd infection, such as an endochitinase, a ß-glucanase, and pathogenesis-related proteins, PR10 and P69G. All these defense proteins were also induced by gentisic acid, a pathogen-induced signal in compatible interactions. The second group of proteins showed no changes at the transcriptional level and included several ribosomal proteins and translation factors, such as the elongation factors 1 and 2 and the translation initiation factor 5-alpha. These results were validated by 2D Western blot, and possible PTMs caused by CEVd infection were detected. Moreover, an interaction between eukaryotic elongation factor 1 and CEVd was observed by 2D Northwestern. The present study provides new protein-related information on the mechanisms of plant resistance to pathogens.


Subject(s)
Gene Expression Regulation, Plant/physiology , Solanum lycopersicum/physiology , Viroids/physiology , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-1/metabolism , Gene Expression Regulation, Plant/drug effects , Gentisates/pharmacology , Solanum lycopersicum/genetics , Solanum lycopersicum/metabolism , Solanum lycopersicum/virology , Plant Diseases , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Polymerase Chain Reaction , Protein Binding , Protein Modification, Translational/drug effects , Protein Modification, Translational/physiology , Proteome/drug effects , Proteome/physiology , RNA, Viral/chemistry , RNA, Viral/metabolism , Reproducibility of Results , Salicylic Acid/pharmacology
10.
Neurotox Res ; 24(2): 139-47, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23325603

ABSTRACT

Previous reports have revealed that reactive oxygen species (ROS) is involved in the development of Alzheimer's disease (AD), and recent studies indicate that free radical-generating systems can regulate amyloid-ß precursor protein (APP) processing. Edaravone is a novel free radical scavenger currently used to reduce cerebral damages after acute cerebral infarction. In the present study, we used SH-SY5Y cells stably transfected with the human "Swedish" APP mutation APP695 (SY5Y-APP695swe) as an in vitro model to investigate the effect of edaravone on APP processing. The result showed that edaravone treatment for 24 h down-regulated ß-amyloid (Aß) production in a dose-dependent manner. Moreover, edaravone modulated APP processing by increasing α-secretase-derived APP fragments and decreasing ß-secretase-derived APP fragments. In addition, the mRNA and protein levels of insulin degrading enzyme (IDE) and neprilysin (NEP), two key Aß degrading enzymes, were not changed after edaravone administration. Taken together, our data suggested that edaravone played an important role in regulating Aß production by enhancing the non-amyloidogenic pathway and inhibiting the amyloidogenic pathway. Thus, edaravone may be potentially useful for treating Alzheimer's disease (AD).


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Antipyrine/analogs & derivatives , Free Radical Scavengers/pharmacology , Protein Modification, Translational/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Antipyrine/pharmacology , Antipyrine/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Edaravone , Free Radical Scavengers/therapeutic use , Humans , Protein Modification, Translational/physiology , Treatment Outcome
11.
J Periodontol ; 84(4): 545-55, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22769443

ABSTRACT

BACKGROUND: Nitric oxide (NO) could be a potential target for the development of new therapeutic approaches to the treatment of periodontal disease because this molecule plays a significant role in the tissue destruction observed in periodontitis. In this study, the authors investigate the effect of kaempferol on the production of NO by murine macrophage-like RAW264.7 cells stimulated with lipopolysaccharide (LPS) from Prevotella intermedia, a pathogen implicated in periodontal disease, and try to determine the underlying mechanisms of action. METHODS: NO production was assayed by measuring the accumulation of nitrite in culture supernatants. Real-time polymerase chain reaction was performed to quantify inducible NO synthase (iNOS) and heme oxygenase-1 (HO-1) mRNA expression. iNOS and HO-1 protein expression and phosphorylation of c-Jun N-terminal kinase and p38 were characterized via immunoblot analysis. Reactive oxygen species (ROS) production was measured using the redox-sensitive fluorescent probe 2',7'-dichlorodihydrofluorescein diacetate. RESULTS: Kaempferol significantly inhibited NO production and expression of iNOS protein in P. intermedia LPS-stimulated RAW246.7 cells without affecting iNOS mRNA expression. Kaempferol upregulated HO-1 expression in LPS-activated cells. Inhibition of HO-1 activity by tin protoporphyrin IX (SnPP) abolished the suppressive effect of kaempferol on NO production. In addition, kaempferol significantly attenuated P. intermedia LPS-induced increase of intracellular ROS, and SnPP blocked this reduction. Treatment with antioxidants downregulated the production of LPS-induced NO. CONCLUSIONS: Kaempferol inhibits NO production and iNOS protein expression in P. intermedia LPS-stimulated RAW264.7 cells at the translational level via HO-1-mediated ROS reduction and could be an efficient modulator of host response in the treatment of periodontal disease.


Subject(s)
Heme Oxygenase-1/physiology , Kaempferols/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Animals , Cell Line , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Prevotella intermedia/metabolism , Protein Modification, Translational/drug effects
12.
Histochem Cell Biol ; 139(3): 501-4, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23229864

ABSTRACT

We previously described the ribopuromyclation method (RPM) to visualize and quantitate translating ribosomes in fixed and permeabilized cells by standard immunofluorescence. RPM is based on puromycylation of nascent chains bound to translating ribosomes followed by detection of puromycylated nascent chains with a puromycin-specific mAb. We now demonstrate that emetine optimally enhances nascent chain puromycylation, and describe a modified RPM protocol for identifying ribosome-bound nascent chains in metabolically inert permeabilized cells.


Subject(s)
Emetine/pharmacology , Protein Modification, Translational/drug effects , Puromycin/metabolism , Cell Membrane Permeability , HeLa Cells , Humans , Ribosomes/drug effects , Ribosomes/metabolism
13.
J Membr Biol ; 245(8): 437-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22886208

ABSTRACT

Gap junctions are key components underpinning multicellularity. They provide cell to cell channel pathways that enable direct intercellular communication and cellular coordination in tissues and organs. The channels are constructed of a family of connexin (Cx) membrane proteins. They oligomerize inside the cell, generating hemichannels (connexons) composed of six subunits arranged around a central channel. After transfer to the plasma membrane, arrays of Cx hemichannels (CxHcs) interact and couple with partners in neighboring attached cells to generate gap junctions. Cx channels have been studied using a range of technical approaches. Short peptides corresponding to sequences in the extra- and intracellular regions of Cxs were used first to generate epitope-specific antibodies that helped studies on the organization and functions of gap junctions. Subsequently, the peptides themselves, especially Gap26 and -27, mimetic peptides derived from each of the two extracellular loops of connexin43 (Cx43), a widely distributed Cx, have been extensively applied to block Cx channels and probe the biology of cell communication. The development of a further series of short peptides mimicking sequences in the intracellular loop, especially the extremity of the intracellular carboxyl tail of Cx43, followed. The primary inhibitory action of the peptidomimetics occurs at CxHcs located at unapposed regions of the cell's plasma membrane, followed by inhibition of cell coupling occurring across gap junctions. CxHcs respond to a range of environmental conditions by increasing their open probability. Peptidomimetics provide a way to block the actions of CxHcs with some selectivity. Furthermore, they are increasingly applied to address the pathological consequences of a range of environmental stresses that are thought to influence Cx channel operation. Cx peptidomimetics show promise as candidates in developing new therapeutic approaches for containing and reversing damage inflicted on CxHcs, especially in hypoxia and ischemia in the heart and in brain functions.


Subject(s)
Cell Membrane/metabolism , Connexins/drug effects , Connexins/metabolism , Gap Junctions/metabolism , Ion Channel Gating , Peptidomimetics/pharmacology , Protein Modification, Translational/drug effects , Animals , Humans
14.
J Neurochem ; 120 Suppl 1: 22-33, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22122190

ABSTRACT

The prescribed drugs for treatment of cognitive deficits in Alzheimer's disease (AD) patients are regarded as symptomatic drugs. Effective disease modifying therapies are not yet prescribed in AD patients. Three major hallmarks of AD (e.g. cholinergic hypofunction, Aß and tau neuropathologies) are closely linked raising the expectation that restoring the cholinergic hypofunction to normal, in particular via selective activation of M1 muscarinic receptors, may alter the onset or progression of AD dementia. This review is focused mainly on modulation of amyloid precursor processing and Aß levels in the brain via cholinergic treatment strategies based on M1 muscarinic agonists versus other cholinergic treatments (e.g. cholinesterase inhibitors prescribed for treatment of AD, M2 antagonists and nicotinic agonists). Advantages and potential drawbacks of these treatment modalities are reviewed versus the notion that due to an elusive etiology of AD, future disease modifiers should address comprehensively most of these AD hallmarks (e.g. Aß pathology, tau and tangle pathologies, as well as the cholinergic hypofunction and cognitive impairments). This major requirement may be fulfilled with M1-selective muscarinic agonists and less with other reviewed cholinergic treatments.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cholinergic Agents/therapeutic use , Protein Modification, Translational , Receptor, Muscarinic M1/metabolism , Alzheimer Disease/drug therapy , Animals , Cholinergic Agents/pharmacology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Disease Models, Animal , Humans , Protein Modification, Translational/drug effects , Protein Modification, Translational/physiology , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/antagonists & inhibitors , Treatment Outcome
15.
J Pharmacol Exp Ther ; 339(1): 210-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21742809

ABSTRACT

Phosphodiesterase 10A (PDE10A) inhibitors have recently been proposed as a new therapy for schizophrenia. The aim of this study was to enhance our understanding of the role of PDE10A inhibitors and potentially identify a clinically useful mechanistic/functional biomarker by using 2-deoxyglucose (2-DG) autoradiography. PDE10A inhibitors papaverine (10 and 40 mg/kg), 6,7-dimethoxy-4-[(3R)-3-(2-quinoxalinyloxy)-1-pyrrolidinyl]quinazoline (PQ-10), (0.16-10 mg/kg), and 2-[{4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)phenoxy}methyl]quinoline (MP-10) (0.16-40 mg/kg) induced region-specific hypermetabolism in the globus pallidus and lateral habenula of C57BL/6 mice. Studies with MP-10 revealed a dose-dependent relative increase in globus pallidus activation, whereas a bell-shaped curve was observed for the lateral habenula. Although the relative increase in 2-DG uptake in the lateral habenula was also characteristic of the D(2) antagonist haloperidol (0.01-0.63 mg/kg), relative 2-DG changes were absent in the globus pallidus. This observation probably is explained by the interaction of PDE10A inhibitors with the D(1) direct pathway as suggested by experiments in combination with the D(1) agonist (±)-6-chloro-7,8-dihydroxy-3-allyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide (SKF-82958) (0.16 mg/kg). The absence of an effect of MP-10 (2.5 mg/kg) on relative glucose metabolism in the globus pallidus and lateral habenula of PDE10A knockout mice confirmed the specificity of the signal induced by PDE10A inhibitors. These studies substantiate the regulatory role of PDE10A in the basal ganglia circuit and as such support the potential of PDE10A inhibitors for treating psychiatric disorders. Moreover, we could differentiate PDE10A inhibitors from haloperidol based on specific patterns of hypermetabolism probably caused by its combined action at both direct and indirect dopaminergic pathways. Finally, these specific changes in brain glucose metabolism may act as a translational biomarker for target engagement in future clinical studies.


Subject(s)
Brain Chemistry/drug effects , Glucose/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Animals , Antimetabolites , Antipsychotic Agents/pharmacology , Autoradiography , Benzazepines/pharmacology , Biomarkers/analysis , Densitometry , Deoxyglucose , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Haloperidol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Papaverine/pharmacology , Phosphoric Diester Hydrolases/genetics , Protein Modification, Translational/drug effects
16.
EMBO J ; 30(17): 3567-80, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21792172

ABSTRACT

In budding yeast, several mRNAs are selectively transported into the daughter cell in an actin-dependent manner by a specialized myosin system, the SHE machinery. With ABP140 mRNA, we now describe the first mRNA that is transported in the opposite direction and localizes to the distal pole of the mother cell, independent of the SHE machinery. Distal pole localization is not observed in mutants devoid of actin cables and can be disrupted by latrunculin A. Furthermore, localization of ABP140 mRNA requires the N-terminal actin-binding domain of Abp140p to be expressed. By replacing the N-terminal localization motif, ABP140 mRNA can be retargeted to different subcellular structures. In addition, accumulation of the mRNA at the distal pole can be prevented by disruption of polysomes. Using the MS2 system, the mRNA was found to associate with actin cables and to follow actin cable dynamics. We therefore propose a model of translational coupling, in which ABP140 mRNA is tethered to actin cables via its nascent protein product and is transported to the distal pole by actin retrograde flow.


Subject(s)
Microfilament Proteins/metabolism , Protein Modification, Translational/physiology , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Biological Transport/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Polyribosomes/drug effects , Polyribosomes/metabolism , Protein Modification, Translational/drug effects , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/drug effects , Thiazolidines/pharmacology
17.
Neuropharmacology ; 61(1-2): 69-79, 2011.
Article in English | MEDLINE | ID: mdl-21392515

ABSTRACT

There remains an urgent need for therapeutic agents that provide improved symptomatic treatment and attenuate disease progression in patients with Alzheimer's disease (AD). 5-HT(4) receptors are widely expressed in those CNS areas which receive substantial cholinergic input and are involved in cognition. The ability of 5-HT(4) receptor agonists to increase acetylcholine (ACh) release and reduce cognitive impairment in both animals and humans has been demonstrated. In addition, 5-HT(4) receptor agonist modulation of levels of the amyloid precursor protein (APP) derived peptides, soluble amyloid precursor protein (sAPPα) and amyloid beta protein (Aß) in the CNS has been reported. In this study, the preclinical properties of three structurally-distinct 5-HT(4) receptor selective agonists, PRX-03140, velusetrag and TD-8954, were studied to assess their potential for symptomatic and disease-modifying benefit in the treatment of AD. All three compounds exhibited high affinity for the rat 5-HT(4) receptor but could be discriminated on the basis of their agonist activity. In cAMP accumulation and sAPPα secretion assays using recombinant HEK293f-5-HT(4(d))-APP(695) cells, velusetrag and TD-8954 were potent, full agonists, relative to 5-HT, whereas PRX-03140 was a partial agonist (intrinsic activity 18%, relative to 5-HT). In a guinea pig colon isolated tissue preparation, TD-8954 exhibited lower intrinsic activity than velusetrag, and PRX-03140 had negligible agonist activity. In the rat Morris water maze (MWM) cognition test, velusetrag and TD-8954 (0.1 mg/kg), but not PRX-03140 (0.03-1 mg/kg), significantly reversed the scopolamine-induced spatial learning deficit via activation of 5-HT(4) receptors. Coadministration of subefficacious doses of the acetylcholinesterase inhibitor (AChEi), donepezil (0.1 mg/kg, i.p.), and either velusetrag or TD-8954 (0.01 mg/kg i.p.) resulted in reversal of the scopolamine-induced cognitive deficit. Pharmacokinetic data indicated that the CNS penetration for all three 5-HT(4) receptor agonists was relatively low. However, the pharmacodynamic-pharmacokinetic relationships in the MWM model for velusetrag and TD-8954 were consistent with their respective receptor pharmacology (binding affinity and intrinsic efficacy) and CNS penetration properties. Collectively, these findings support a potential role for potent and efficacious 5-HT(4) receptor agonists in the treatment of AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cognition/physiology , Protein Modification, Translational/physiology , Receptors, Serotonin, 5-HT4/physiology , Serotonin 5-HT4 Receptor Agonists/pharmacology , Animals , Cognition/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , HEK293 Cells , Humans , Male , Protein Binding/physiology , Protein Modification, Translational/drug effects , Rats , Rats, Sprague-Dawley , Serotonin 5-HT4 Receptor Agonists/pharmacokinetics
18.
Neurotoxicology ; 32(1): 58-67, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20970449

ABSTRACT

The blood-brain barrier (BBB) is essential for central nervous system (CNS) normal function. It is formed by endothelial cells with special characteristics, which confer the BBB with low permeability and high transendothelial electrical resistance (TEER). We previously demonstrated that malathion and lead, two neurotoxicants widely present in the environment, decrease TEER and increase permeability in in vitro models of the BBB. In this study we assessed tight junction disruption at the protein and gene expression levels using a rat brain microvascular endothelial cell line (RBE4) exposed to lead acetate at 10(-5)M and 10(-6)M, malathion at 10(-5)M, malaoxon at 10(-6)M, and their combinations. Cells were incubated with treatments for 2h, 4h, 8h, 16h, and 24h periods. Immunoblotting assessments demonstrated that protein levels of tight junction proteins occludin and claudin 5, and scaffold proteins ZO1 and ZO2 were decreased after treatments. Gene expression determinations did not correlate with the decreases in protein, indicating that the effects on these proteins were post-translational.


Subject(s)
Blood-Brain Barrier/metabolism , Claudins/metabolism , Malathion/toxicity , Membrane Proteins/metabolism , Organometallic Compounds/toxicity , Phosphoproteins/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Line , Claudin-5 , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Occludin , Protein Modification, Translational/drug effects , Rats , Zonula Occludens-1 Protein , Zonula Occludens-2 Protein
19.
Toxicol Lett ; 196(1): 51-5, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20382208

ABSTRACT

The present study investigated in mice brain, the time course of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity on the expression of translational control proteins. Mice received intraperitoneal injections of MPTP (30 mg/kg/day) for 5 days and were sacrificed 1, 2, 3, 4 and 7 days after the last injection. The results, obtained by western blot, indicated that MPTP produced an alteration of the expression of proteins involved in the mTOR anti-apoptotic way and the PKR pro-apoptotic pathway of translational control especially in striatum and frontal cortex of mice. These disturbances were associated with a great activation of PKR in hippocampus at D9, the time point corresponding to maximal translational control alterations. Furthermore, whereas no modification of translational control protein expression was observed in mice substantia nigra after western blot procedure, immunofluorescent labeling revealed, in this target region of the toxin MPTP, a decrease of the expression of phospho-mTOR and a great activation of the phosphorylated form of PKR, marker of pathogenesis.


Subject(s)
Brain/metabolism , MPTP Poisoning/metabolism , Neurotoxins/toxicity , Protein Modification, Translational/drug effects , Animals , Brain/drug effects , Brain/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases , Time , Toxicity Tests , eIF-2 Kinase/metabolism
20.
J Neurochem ; 113(5): 1133-49, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345768

ABSTRACT

The bioactivity of endothelin-1 (ET-1) has been suggested in the development of CNS diseases, including disturbance of water homeostasis and blood-brain barrier integrity. Recent studies suggest that hypoxic/ischemic injury of the brain induces release of ET-1, behaving through a G-protein coupled ET receptor family. The deleterious effects of ET-1 on astrocytes may aggravate brain inflammation. Increased plasma levels of matrix metalloproteinases (MMPs), in particular MMP-9, have been observed in patients with neuroinflammatory disorders. However, the detailed mechanisms underlying ET-1-induced MMP-9 expression remain unknown. In this study, the data obtained with zymographic, western blotting, real-time PCR, and immunofluorescent staining analyses showed that ET-1-induced MMP-9 expression was mediated through an ET(B)-dependent transcriptional activation. Engagement of G(i/o)- and G(q)-coupled ET(B) receptor by ET-1 led to activation of p42/p44 MAPK and then activated transcription factors including Ets-like kinase, nuclear factor-kappa B, and activator protein-1 (c-Jun/c-Fos). These activated transcription factors translocated into nucleus and bound to their corresponding binding sites in MMP-9 promoter, thereby turning on MMP-9 gene transcription. Eventually, up-regulation of MMP-9 by ET-1 enhanced the migration of astrocytes. Taken together, these results suggested that in astrocytes, activation of Ets-like kinase, nuclear factor-kappa B, and activator protein-1 by ET(B)-dependent p42/p44 MAPK signaling is necessary for ET-1-induced MMP-9 gene up-regulation. Understanding the mechanisms of MMP-9 expression and functional changes regulated by ET-1/ET(B) system on astrocytes may provide rational therapeutic interventions for brain injury associated with increased MMP-9 expression.


Subject(s)
Astrocytes/drug effects , Astrocytes/enzymology , Brain/cytology , Cell Movement/drug effects , Endothelin-1/pharmacology , Matrix Metalloproteinase 9/biosynthesis , Animals , Blotting, Western , Brain/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 9/genetics , Plasmids/genetics , Promoter Regions, Genetic/genetics , Protein Modification, Translational/drug effects , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Transcriptional Activation/drug effects , Transfection , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...