Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Brain ; 140(5): 1280-1299, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28334907

ABSTRACT

Ataxin-3, the disease protein in Machado-Joseph disease, is known to be proteolytically modified by various enzymes including two major families of proteases, caspases and calpains. This processing results in the generation of toxic fragments of the polyglutamine-expanded protein. Although various approaches were undertaken to identify cleavage sites within ataxin-3 and to evaluate the impact of fragments on the molecular pathogenesis of Machado-Joseph disease, calpain-mediated cleavage of the disease protein and the localization of cleavage sites remained unclear. Here, we report on the first precise localization of calpain cleavage sites in ataxin-3 and on the characterization of the resulting breakdown products. After confirming the occurrence of calpain-derived fragmentation of ataxin-3 in patient-derived cell lines and post-mortem brain tissue, we combined in silico prediction tools, western blot analysis, mass spectrometry, and peptide overlay assays to identify calpain cleavage sites. We found that ataxin-3 is primarily cleaved at two sites, namely at amino acid positions D208 and S256 and mutating amino acids at both cleavage sites to tryptophan nearly abolished ataxin-3 fragmentation. Furthermore, analysis of calpain cleavage-derived fragments showed distinct aggregation propensities and toxicities of C-terminal polyglutamine-containing breakdown products. Our data elucidate the important role of ataxin-3 proteolysis in the pathogenesis of Machado-Joseph disease and further emphasize the relevance of targeting this disease pathway as a treatment strategy in neurodegenerative disorders.


Subject(s)
Ataxin-3/metabolism , Calpain/metabolism , Machado-Joseph Disease/metabolism , Brain/metabolism , Cells, Cultured , Combinatorial Chemistry Techniques , Computer Simulation , Humans , Induced Pluripotent Stem Cells/metabolism , Peptide Hydrolases/metabolism , Protein Aggregation, Pathological/metabolism , Transfection
2.
J Biol Chem ; 287(2): 1279-89, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22110140

ABSTRACT

Huntington disease (HD), a fatal neurodegenerative disorder, is caused by a lengthening of the polyglutamine tract in the huntingtin (Htt) protein. Despite considerable effort, thus far there is no cure or treatment available for the disorder. Using the approach of tandem affinity purification we recently discovered that prothymosin-α (ProTα), a small highly acidic protein, interacts with mutant Htt (mHtt). This was confirmed by co-immunoprecipitation and a glutathione S-transferase (GST) pull-down assay. Overexpression of ProTα remarkably reduced mHtt-induced cytotoxicity in both non-neuronal and neuronal cell models expressing N-terminal mHtt fragments, whereas knockdown of ProTα expression in the cells enhanced mHtt-caused cell death. Deletion of the central acidic domain of ProTα abolished not only its interaction with mHtt but also its protective effect on mHtt-caused cytotoxicity. Additionally, overexpression of ProTα inhibited caspase-3 activation but enhanced aggregation of mHtt. Furthermore, when added to cultured cells expressing mHtt, the purified recombinant ProTα protein not only entered the cells but it also significantly suppressed the mHtt-caused cytotoxicity. Taken together, these data suggest that ProTα might be a novel therapeutic target for treating HD and other polyglutamine expansion disorders.


Subject(s)
Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Thymosin/analogs & derivatives , Amino Acid Sequence , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/drug effects , Enzyme Activation/drug effects , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Protein Binding , Protein Precursors/genetics , Protein Precursors/pharmacology , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Deletion , Thymosin/genetics , Thymosin/metabolism , Thymosin/pharmacology
3.
Mol Cell Proteomics ; 10(1): M110.001172, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876399

ABSTRACT

Mutations in human leucine-rich repeat kinase 2 (Lrrk2), a protein of yet unknown function, are linked to Parkinson's disease caused by degeneration of midbrain dopaminergic neurons. The protein comprises several domains including a GTPase and a kinase domain both affected by several pathogenic mutations. To elucidate the molecular interaction network of endogenous Lrrk2 under stoichiometric constraints, we applied QUICK (quantitative immunoprecipitation combined with knockdown) in NIH3T3 cells. The identified interactome reveals actin isoforms as well as actin-associated proteins involved in actin filament assembly, organization, rearrangement, and maintenance, suggesting that the biological function of Lrrk2 is linked to cytoskeletal dynamics. In fact, we demonstrate Lrrk2 de novo binding to F-actin and its ability to modulate its assembly in vitro. When tested in intact cells, knockdown of Lrrk2 causes morphological alterations in NIH3T3 cells. In developing dopaminergic midbrain primary neurons, Lrrk2 knockdown results in shortened neurite processes, indicating a physiological role of Lrrk2 in cytoskeletal organization and dynamics of dopaminergic neurons. Hence, our results demonstrate that molecular interactions as well as the physiological function of Lrrk2 are closely related to the organization of the actin-based cytoskeleton, a crucial feature of neuronal development and neuron function.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Immunoprecipitation/methods , Protein Serine-Threonine Kinases/metabolism , Animals , Dopamine/metabolism , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , HEK293 Cells , Humans , Lentivirus/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mesencephalon/enzymology , Mice , NIH 3T3 Cells , Neurites/metabolism , Polymerization , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Reproducibility of Results , Signal Transduction
4.
Proteomics ; 12(12): 2060-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22623107

ABSTRACT

Huntington's disease (HD) is caused by a CAG triplet repeat expansion in exon 1 of the Huntingtin (Htt) gene, encoding an abnormal expanded polyglutamine (polyQ) tract that confers toxicity to the mutant Htt (mHtt) protein. Recent data suggest that posttranslational modifications of mHtt modulate its cytotoxicity. To further understand the cytotoxic mechanisms of mHtt, we have generated HEK293 cell models stably expressing Strep- and FLAG-tagged Htt containing either 19Q (wild-type Htt), 55Q (mHtt), or 94Q (mHtt) repeats. Following tandem affinity purification, the tagged Htt and associated proteins were subjected to tandem mass spectrometry or 2D nano-LC tandem mass spectrometry and several novel modification sites of mHtt containing 55Q or 94Q were identified. These were phosphorylation sites located at Ser431 and Ser432, and ubiquitination site located at Lys444. The two phosphorylation sites were confirmed by Western blot analysis using phosphorylation site-specific antibodies. In addition, prevention of phosphorylation at the two serine sites altered mHtt toxicity and accumulation. These modifications of mHtt may provide novel therapeutic targets for effective treatment of the disorder.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Chromatography, Affinity , HEK293 Cells , Humans , Huntingtin Protein , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Phosphorylation , Ubiquitination
5.
J Neurochem ; 123(3): 437-46, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22891683

ABSTRACT

Huntington's disease (HD) is a devastating neurodegenerative disorder caused by an expansion of CAG trinucleotide repeats encoding for polyglutamine (polyQ) in the huntingtin (Htt) gene. Despite considerable effort, the mechanisms underlying the toxicity of the mutated Htt protein remains largely uncertain. To identify novel therapeutic targets, we recently employed the approach of tandem affinity purification and discovered that calretinin (Cr), a member of the EF-hand family of calcium-binding proteins, is preferentially associated with mHtt, although it also interacts with wild-type Htt. These observations were supported by coimmunoprecipitation and by colocalization of Cr with mHtt in neuronal cultures. Over- expression of Cr reduced mHtt-caused cytotoxicity in both non-neuronal and neuronal cell models of HD, whereas knockdown of Cr expression in the cells enhanced mHtt-caused neuronal cell death. In addition, over-expression of Cr was also associated with reduction of intracellular free calcium and activation of Akt. These results suggest that Cr may be a potential therapeutic target for treatment of HD.


Subject(s)
Down-Regulation/genetics , Huntington Disease/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , S100 Calcium Binding Protein G/metabolism , Animals , Calbindin 2 , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Disease Models, Animal , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/prevention & control , Male , Mice , Mice, Neurologic Mutants , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , S100 Calcium Binding Protein G/genetics , S100 Calcium Binding Protein G/physiology
6.
BMC Dev Biol ; 11: 27, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21586122

ABSTRACT

BACKGROUND: Neurogenesis control and the prevention of premature differentiation in the vertebrate embryo are crucial processes, allowing the formation of late-born cell types and ensuring the correct shape and cytoarchitecture of the brain. Members of the Hairy/Enhancer of Split (Hairy/E(spl)) family of bHLH-Orange transcription factors, such as zebrafish Her3, 5, 9 and 11, are implicated in the local inhibition of neurogenesis to maintain progenitor pools within the early neural plate. To better understand how these factors exert their inhibitory function, we aimed to isolate some of their functional interactors. RESULTS: We used a yeast two-hybrid screen with Her5 as bait and recovered a novel zebrafish Hairy/E(spl) factor--Her8a. Using phylogenetic and synteny analyses, we demonstrate that her8a evolved from an ancient duplicate of Hes6 that was recently lost in the mammalian lineage. We show that her8a is expressed across the mid- and anterior hindbrain from the start of segmentation. Through knockdown and misexpression experiments, we demonstrate that Her8a is a negative regulator of neurogenesis and plays an essential role in generating progenitor pools within rhombomeres 2 and 4--a role resembling that of Her3. Her8a co-purifies with Her3, suggesting that Her8a-Her3 heterodimers may be relevant in this domain of the neural plate, where both proteins are co-expressed. Finally, we demonstrate that her8a expression is independent of Notch signaling at the early neural plate stage but that SoxB factors play a role in its expression, linking patterning information to neurogenesis control. Overall, the regulation and function of Her8a differ strikingly from those of its closest relative in other vertebrates--the Hes6-like proteins. CONCLUSIONS: Our results characterize the phylogeny, expression and functional interactions involving a new Her factor, Her8a, and highlight the complex interplay of E(spl) proteins that generates the neurogenesis pattern of the zebrafish early neural plate.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Repressor Proteins/metabolism , Rhombencephalon/embryology , Zebrafish Proteins/metabolism , Zebrafish/anatomy & histology , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Morphogenesis/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/genetics , Phylogeny , Protein Binding , Protein Multimerization , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Rhombencephalon/cytology , Signal Transduction/physiology , Two-Hybrid System Techniques , Zebrafish Proteins/chemistry , Zebrafish Proteins/classification , Zebrafish Proteins/genetics
7.
Mol Cell Proteomics ; 7(7): 1349-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18436526

ABSTRACT

Glial cells support neuronal survival and function by secreting neurotrophic cytokines. Retinal Mueller glial cells (RMGs) support retinal neurons, especially photoreceptors. These highly light-sensitive sensory neurons receive vision, and their death results in blinding diseases. It has been proposed that RMGs release factors that support photoreceptor survival, but the nature of these factors remains to be elucidated. To discover such neurotrophic factors, we developed an integrated work flow toward systematic identification of neuroprotective proteins, which are, like most cytokines, expressed only in minute amounts. This strategy can be generally applied to identify secreted bioactive molecules from any body fluid once a recipient cell for this activity is known. Toward this goal we first isolated conditioned medium (CM) from primary porcine RMGs cultured in vitro and tested for survival-promoting activity using primary photoreceptors. We then developed a large scale, microplate-based cellular high content assay that allows rapid assessment of primary photoreceptor survival concomitant with biological activity in vitro. The enrichment strategy of bioactive proteins toward their identification consists of several fractionation steps combined with tests for biological function. Here we combined 1) size fractionation, 2) ion exchange chromatography, 3) reverse phase liquid chromatography, and 4) mass spectrometry (Q-TOF MS/MS or MALDI MS/MS) for protein identification. As a result of this integrated work flow, the insulin-like growth factor-binding proteins IGFBP5 and IGFBP7 and connective tissue growth factor (CTGF) were identified as likely candidates. Cloning and stable expression of these three candidate factors in HEK293 cells produced conditioned medium enriched for either one of the factors. IGFBP5 and CTGF, but not IGFBP7, significantly increased photoreceptor survival when secreted from HEK293 cells and when added to the original RMG-CM. This indicates that the survival-promoting activity in RMG-CM is multifactorial with IGFBP5 and CTGF as an integral part of this activity.


Subject(s)
Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/isolation & purification , Paracrine Communication , Proteomics/methods , Algorithms , Animals , Anion Exchange Resins/metabolism , Cell Fractionation/methods , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Eye/innervation , Feasibility Studies , Humans , Nerve Tissue Proteins/analysis , Neuroglia/metabolism , Neuroprotective Agents/analysis , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Photoreceptor Cells/drug effects , Photoreceptor Cells/physiology , Swine
8.
iScience ; 23(12): 101797, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33299968

ABSTRACT

PINK1 loss-of-function mutations cause early onset Parkinson disease. PINK1-Parkin mediated mitophagy has been well studied, but the relevance of the endogenous process in the brain is debated. Here, the absence of PINK1 in human dopaminergic neurons inhibits ionophore-induced mitophagy and reduces mitochondrial membrane potential. Compensatory, mitochondrial renewal maintains mitochondrial morphology and protects the respiratory chain. This is paralleled by metabolic changes, including inhibition of the TCA cycle enzyme mAconitase, accumulation of NAD+, and metabolite depletion. Loss of PINK1 disrupts dopamine metabolism by critically affecting its synthesis and uptake. The mechanism involves steering of key amino acids toward energy production rather than neurotransmitter metabolism and involves cofactors related to the vitamin B6 salvage pathway identified using unbiased multi-omics approaches. We propose that reduction of mitochondrial membrane potential that cannot be controlled by PINK1 signaling initiates metabolic compensation that has neurometabolic consequences relevant to Parkinson disease.

9.
Front Mol Neurosci ; 11: 64, 2018.
Article in English | MEDLINE | ID: mdl-29541021

ABSTRACT

Mutations in leucine-rich repeat kinase 2 gene (LRRK2) are associated with familial and sporadic Parkinson's disease (PD). LRRK2 is a complex protein that consists of multiple domains, including 13 putative armadillo-type repeats at the N-terminus. In this study, we analyzed the functional and molecular consequences of a novel variant, E193K, identified in an Italian family. E193K substitution does not influence LRRK2 kinase activity. Instead it affects LRRK2 biochemical properties, such as phosphorylation at Ser935 and affinity for 14-3-3ε. Primary fibroblasts obtained from an E193K carrier demonstrated increased cellular toxicity and abnormal mitochondrial fission upon 1-methyl-4-phenylpyridinium treatment. We found that E193K alters LRRK2 binding to DRP1, a crucial mediator of mitochondrial fission. Our data support a role for LRRK2 as a scaffolding protein influencing mitochondrial fission.

10.
Methods Mol Biol ; 1188: 177-90, 2014.
Article in English | MEDLINE | ID: mdl-25059612

ABSTRACT

Pull-downs based on tag fusion proteins as well as immunoprecipitations (IP) are widely used methods to analyze protein interactions. Selectivity and specificity of both methods are compromised by nonspecific binding to the capture agent or carrier beads thereby generating false positives. Here, we provide a method combining stable isotope labeling of amino acids in cell culture (SILAC) with affinity purification, coupled to quantitative tandem mass spectrometry. It permits the analysis of protein interactions with high sensitivity, while being able to discriminate contaminants and nonspecific binders. Besides pruning out contaminants, high-resolution MS data combined with quantitative proteomics software allow the comparative analysis of protein interaction patterns of different protein variants, for example mutated versus normal protein variant or of regulatory changes in a given protein complex due to different states of activity.


Subject(s)
Amino Acids/chemistry , Isotope Labeling/methods , Proteins/chemistry , Proteins/metabolism , Proteomics/methods , Analytic Sample Preparation Methods , Cell Death , HEK293 Cells , Humans , Mass Spectrometry , Proteins/isolation & purification
11.
J Neurosci Methods ; 214(1): 62-8, 2013 Mar 30.
Article in English | MEDLINE | ID: mdl-23318290

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. LRRK2 kinase activity is required for toxicity in neuronal cell cultures suggesting that selective kinase inhibitors may prevent neurodegeneration in patients. Directly monitoring LRRK2 activity in cells would be advantageous for the development of small molecule LRRK2 inhibitors. Here, we demonstrate that a monoclonal anti-LRRK2 antibody directed against the activation segment binds less efficiently to native LRRK2 protein in the presence of ATP-competitive LRRK2 inhibitors. Since kinase inhibitors prevent autophosphorylation and refolding of the activation segment, we hypothesize that the antibody preferentially binds to the active conformation of LRRK2 under native conditions.


Subject(s)
Antibodies, Monoclonal/metabolism , Epitopes/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigen-Antibody Reactions , Benzodiazepinones/pharmacology , Binding, Competitive , Enzyme Activation , Epitopes/chemistry , HEK293 Cells , Humans , Immunoprecipitation , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Parkinson Disease/enzymology , Parkinson Disease/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Pyrimidines/pharmacology , Recombinant Proteins/antagonists & inhibitors , Swiss 3T3 Cells
12.
Dev Cell ; 21(3): 479-91, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21920314

ABSTRACT

The Drosophila Discs large (Dlg) scaffolding protein acts as a tumor suppressor regulating basolateral epithelial polarity and proliferation. In mammals, four Dlg homologs have been identified; however, their functions in cell polarity remain poorly understood. Here, we demonstrate that the X-linked mental retardation gene product Dlg3 contributes to apical-basal polarity and epithelial junction formation in mouse organizer tissues, as well as to planar cell polarity in the inner ear. We purified complexes associated with Dlg3 in polarized epithelial cells, including proteins regulating directed trafficking and tight junction formation. Remarkably, of the four Dlg family members, Dlg3 exerts a distinct function by recruiting the ubiquitin ligases Nedd4 and Nedd4-2 through its PPxY motifs. We found that these interactions are required for Dlg3 monoubiquitination, apical membrane recruitment, and tight junction consolidation. Our findings reveal an unexpected evolutionary diversification of the vertebrate Dlg family in basolateral epithelium formation.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Cell Polarity/physiology , Ear, Inner/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Protein Transport , Ubiquitin-Protein Ligases/genetics
13.
J Clin Invest ; 121(6): 2169-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21606596

ABSTRACT

The mutations that cause Leber congenital amaurosis (LCA) lead to photoreceptor cell death at an early age, causing childhood blindness. To unravel the molecular basis of LCA, we analyzed how mutations in LCA5 affect the connectivity of the encoded protein lebercilin at the interactome level. In photoreceptors, lebercilin is uniquely localized at the cilium that bridges the inner and outer segments. Using a generally applicable affinity proteomics approach, we showed that lebercilin specifically interacted with the intraflagellar transport (IFT) machinery in HEK293T cells. This interaction disappeared when 2 human LCA-associated lebercilin mutations were introduced, implicating a specific disruption of IFT-dependent protein transport, an evolutionarily conserved basic mechanism found in all cilia. Lca5 inactivation in mice led to partial displacement of opsins and light-induced translocation of arrestin from photoreceptor outer segments. This was consistent with a defect in IFT at the connecting cilium, leading to failure of proper outer segment formation and subsequent photoreceptor degeneration. These data suggest that lebercilin functions as an integral element of selective protein transport through photoreceptor cilia and provide a molecular demonstration that disrupted IFT can lead to LCA.


Subject(s)
Eye Proteins/physiology , Leber Congenital Amaurosis/physiopathology , Microtubule-Associated Proteins/physiology , Photoreceptor Connecting Cilium/physiology , Protein Transport/physiology , Animals , Arrestins/metabolism , Cell Line , Disease Models, Animal , Eye Proteins/genetics , Humans , Leber Congenital Amaurosis/genetics , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Multiprotein Complexes , Opsins/metabolism , Protein Interaction Mapping , Protein Transport/genetics , Recombinant Fusion Proteins/physiology , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/pathology , Vision, Ocular/physiology
14.
Invest Ophthalmol Vis Sci ; 51(1): 79-88, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19696174

ABSTRACT

PURPOSE: SNPs in chromosomal region 10q26 harboring PLEKHA1, ARMS2, and Htra1 showed the strongest association with age-related macular degeneration. Recent evidence suggests that in patients homozygous for the risk allele, the lack of synthesis of the poorly characterized ARMS2 is causative of this disorder. The present study was undertaken to gain an understanding of the genuine (patho)physiological role of this protein. METHODS: ARMS2-interacting proteins were identified by using a yeast two-hybrid system and validated by coprecipitation. Immunofluorescence was applied to reveal the localization of ARMS2 in transfected cells and in human eyes. Western blot analyses were performed on extra- and intracellular fractions of ARMS2-expressing cells to demonstrate the secretion of ARMS2. RESULTS: Contrary to previous reports, this study showed that ARMS2 is a secreted protein that binds several matrix proteins. Notably, ARMS2 directly interacts with fibulin-6 (hemicentin-1). Mutations in the fibulin-6 gene have been demonstrated to cause familial AMD. ARMS2 also interacts with further extracellular proteins, several of which have been implicated in macular dystrophies. Although ARMS2 apparently lacks any classic targeting sequence, it is translocated to the endoplasmic reticulum in cultured cells before secretion. ARMS2 is mostly confined to choroid pillars in human eyes, representing a part of extracellular matrix and corresponding to the principal sites of drusen formation. CONCLUSIONS: The pivotal role of the extracellular matrix in the progression of AMD is underlined by the abnormal deposition of extracellular debris in the macula, observed frequently in affected individuals. The results have shown that ARMS2 may be necessary for proper matrix function.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Macular Degeneration/metabolism , Proteins/physiology , Aged , Animals , Antibodies, Monoclonal , Blotting, Western , Calcium-Binding Proteins/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunoglobulins/metabolism , Macular Degeneration/genetics , Male , Peptide Fragments , Plasmids , Protein Interaction Mapping , Rats , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Two-Hybrid System Techniques
15.
Glia ; 55(10): 1061-73, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17551930

ABSTRACT

Neurosteroids, such as progesterone, influence central nervous system development and function by regulating a broad spectrum of physiological processes. Here, we investigated membrane-initiated actions of progesterone in the retina and identified the membrane-associated progesterone receptor component 1 (PGRMC1). We found PGRMC1 expressed mainly in retinal Muller glia (RMG) and retinal pigment epithelium, and localized uniquely to microsomal and plasma membrane fractions. In RMG, membrane-impermeable progesterone conjugate induced calcium influx and subsequent phosphatidylinositol 3-kinase-mediated phosphorylation of PKC and ERK-1/2. Induction by progesterone also led to PKC-dependent activation of VEGF gene expression and protein synthesis, suggesting a contribution of membrane-initiated hormone effects to VEGF induced neovascularization within retina.


Subject(s)
Calcium Signaling/physiology , Gene Expression Regulation/physiology , Neuroglia/metabolism , Progesterone/physiology , Retina/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Homeostasis/physiology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neuroglia/cytology , Neuroglia/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Progesterone/pharmacology , Protein Kinase C/drug effects , Protein Kinase C/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Progesterone/drug effects , Receptors, Progesterone/metabolism , Retina/cytology , Retina/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sus scrofa
16.
Mol Cell Proteomics ; 5(11): 2185-200, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16917120

ABSTRACT

One major problem concerning the electrophoresis of mitochondria is the heterogeneity of mitochondrial appearance especially under pathological conditions. We show here the use of zone electrophoresis in a free flow electrophoresis device (ZE-FFE) as an analytical sensor to discriminate between different yeast mitochondrial populations. Impairment of the structural properties of the organelles by hyperosmotic stress resulted in broad separation profiles. Conversely untreated mitochondria gave rise to homogeneous populations reflected by sharp separation profiles. Yeast mitochondria with altered respiratory activity accompanied by a different outer membrane proteome composition could be discriminated based on electrophoretic deflection. Proteolysis of the mitochondrial surface proteome and the deletion of a single major protein species of the mitochondrial outer membrane altered the ZE-FFE deflection of these organelles. To demonstrate the usefulness of ZE-FFE for the analysis of mitochondria associated with pathological processes, we analyzed mitochondrial fractions from an apoptotic yeast strain. The cdc48(S565G) strain carries a mutation in the CDC48 gene that is an essential participant in the endoplasmic reticulum-associated protein degradation pathway. Mutant cells accumulate polyubiquitinated proteins in microsomal and mitochondrial extracts. Subsequent ZE-FFE characterization could distinguish a mitochondrial subfraction specifically enriched with polyubiquitinated proteins from the majority of non-affected mitochondria. This result demonstrates that ZE-FFE may give important information on the specific properties of subpopulations of a mitochondrial preparation allowing a further detailed functional analysis.


Subject(s)
Electrophoresis/instrumentation , Mitochondria/chemistry , Mitochondrial Proteins/analysis , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases , Cell Cycle Proteins/genetics , Glucose/chemistry , Lactic Acid/chemistry , Mitochondria/ultrastructure , Mitochondrial Membranes/chemistry , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL