Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(7)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062128

ABSTRACT

Pathological mutations in the LRRK2 gene are the major genetic cause of Parkinson's disease (PD). Although several animal models with either LRRK2 down- or over-expression have been developed, the physiological function of LRRK2 remains elusive. LRRK2 is constitutively expressed in various tissues including neurons and glial cells, but importantly, it is expressed at low levels in dopaminergic neurons, further contributing to the cryptic function of LRRK2. Significant levels of LRRK2 protein and mRNA have been detected in peripheral blood mononuclear cells, lymph nodes, the spleen, and primary microglia, strongly suggesting the contribution of inflammatory cells to neuronal degeneration. In this research article, using Drosophila LRRK2 models, we were able to demonstrate a significant contribution of glial cells to the LRRK2 pathological phenotype. Furthermore, in Drosophila, neurodegeneration is associated with a significant and important increase in specific inflammatory peptides. Finally, levetiracetam, a compound widely used in human therapy to treat epilepsy, was able to rescue both neuronal degeneration and neuroinflammation.

2.
Front Immunol ; 15: 1360068, 2024.
Article in English | MEDLINE | ID: mdl-38596666

ABSTRACT

The complex interplay between genetic and environmental factors is considered the cause of neurodegenerative diseases including Parkinson's disease (PD) and Amyotrophic Lateral Sclerosis (ALS). Among the environmental factors, toxins produced by cyanobacteria have received much attention due to the significant increase in cyanobacteria growth worldwide. In particular, L-BMAA toxin, produced by diverse taxa of cyanobacteria, dinoflagellates and diatoms, has been extensively correlated to neurodegeneration. The molecular mechanism of L-BMAA neurotoxicity is still cryptic and far from being understood. In this research article, we have investigated the molecular pathways altered by L-BMAA exposure in cell systems, highlighting a significant increase in specific stress pathways and an impairment in autophagic processes. Interestingly, these changes lead to the accumulation of both α-synuclein and TDP43, which are correlated with PD and ALS proteinopathy, respectively. Finally, we were able to demonstrate specific alterations of TDP43 WT or pathological mutants with respect to protein accumulation, aggregation and cytoplasmic translocation, some of the typical features of both sporadic and familial ALS.


Subject(s)
Amino Acids, Diamino , Amyotrophic Lateral Sclerosis , Cyanobacteria , Parkinson Disease , Humans , Amyotrophic Lateral Sclerosis/pathology , alpha-Synuclein , Cyanobacteria Toxins , Amino Acids, Diamino/toxicity
3.
Int J Mol Sci ; 24(16)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37628835

ABSTRACT

Pathological mutations in leucine-rich repeat kinase 2 (LRRK2) gene are the major genetic cause of Parkinson's disease (PD). Multiple lines of evidence link LRRK2 to the control of vesicle dynamics through phosphorylation of a subset of RAB proteins. However, the molecular mechanisms underlying these processes are not fully elucidated. We have previously demonstrated that LRRK2 increases the exocyst complex assembly by Sec8 interaction, one of the eight members of the exocyst complex, and that Sec8 over-expression mitigates the LRRK2 pathological effect in PC12 cells. Here, we extend this analysis using LRRK2 drosophila models and show that the LRRK2-dependent exocyst complex assembly increase is downstream of RAB phosphorylation. Moreover, exocyst complex inhibition rescues mutant LRRK2 pathogenic phenotype in cellular and drosophila models. Finally, prolonged exocyst inhibition leads to a significant reduction in the LRRK2 protein level, overall supporting the role of the exocyst complex in the LRRK2 pathway. Taken together, our study suggests that modulation of the exocyst complex may represent a novel therapeutic target for PD.


Subject(s)
Blister , Parkinson Disease , Animals , Rats , Cytoplasm , Phosphorylation , Drosophila , Exocytosis , Parkinson Disease/genetics
4.
J Cell Biol ; 221(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35266954

ABSTRACT

Missense mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common cause of familial Parkinson's disease (PD); however, pathways regulating LRRK2 subcellular localization, function, and turnover are not fully defined. We performed quantitative mass spectrometry-based interactome studies to identify 48 novel LRRK2 interactors, including the microtubule-associated E3 ubiquitin ligase TRIM1 (tripartite motif family 1). TRIM1 recruits LRRK2 to the microtubule cytoskeleton for ubiquitination and proteasomal degradation by binding LRRK2911-919, a nine amino acid segment within a flexible interdomain region (LRRK2853-981), which we designate the "regulatory loop" (RL). Phosphorylation of LRRK2 Ser910/Ser935 within LRRK2 RL influences LRRK2's association with cytoplasmic 14-3-3 versus microtubule-bound TRIM1. Association with TRIM1 modulates LRRK2's interaction with Rab29 and prevents upregulation of LRRK2 kinase activity by Rab29 in an E3-ligase-dependent manner. Finally, TRIM1 rescues neurite outgrowth deficits caused by PD-driving mutant LRRK2 G2019S. Our data suggest that TRIM1 is a critical regulator of LRRK2, controlling its degradation, localization, binding partners, kinase activity, and cytotoxicity.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease , Protein Serine-Threonine Kinases , Tripartite Motif Proteins , Cytoskeleton , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Microtubule-Associated Proteins , Microtubules , Mutation , Parkinson Disease/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Transcription Factors , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , rab GTP-Binding Proteins/metabolism
5.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445429

ABSTRACT

The prevalence of neurodegenerative disease (ND) is increasing, partly owing to extensions in lifespan, with a larger percentage of members living to an older age, but the ND aetiology and pathogenesis are not fully understood, and effective treatments are still lacking. Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis are generally thought to progress as a consequence of genetic susceptibility and environmental influences. Up to now, several environmental triggers have been associated with NDs, and recent studies suggest that some cyanotoxins, produced by cyanobacteria and acting through a variety of molecular mechanisms, are highly neurotoxic, although their roles in neuropathy and particularly in NDs are still controversial. In this review, we summarize the most relevant and recent evidence that points at cyanotoxins as environmental triggers in NDs development.


Subject(s)
Bacterial Toxins/toxicity , Cyanobacteria/pathogenicity , Neurodegenerative Diseases/etiology , Animals , Cyanobacteria/metabolism , Genetic Predisposition to Disease , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/microbiology
6.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299248

ABSTRACT

Parkinson's disease (PD) is a complex and progressive neurodegenerative disorder with a prevalence of approximately 0.5-1% among those aged 65-70 years. Although most of its clinical manifestations are due to a loss of dopaminergic neurons, the PD etiology is largely unknown. PD is caused by a combination of genetic and environmental factors, and the exact interplay between genes and the environment is still debated. Several biological processes have been implicated in PD, including mitochondrial or lysosomal dysfunctions, alteration in protein clearance, and neuroinflammation, but a common molecular mechanism connecting the different cellular alterations remains incompletely understood. Accumulating evidence underlines a significant role of lipids in the pathological pathways leading to PD. Beside the well-described lipid alteration in idiopathic PD, this review summarizes the several lipid alterations observed in experimental models expressing PD-related genes and suggests a possible scenario in relationship to the molecular mechanisms of neuronal toxicity. PD could be considered a lipid-induced proteinopathy, where alteration in lipid composition or metabolism could induce protein alteration-for instance, alpha-synuclein accumulation-and finally neuronal death.


Subject(s)
Lipid Metabolism/genetics , Lipids/physiology , Parkinson Disease/genetics , Dopaminergic Neurons/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Nerve Degeneration/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
7.
Cells ; 10(2)2021 01 20.
Article in English | MEDLINE | ID: mdl-33498474

ABSTRACT

Mutations in LRRK2 play a critical role in both familial and sporadic Parkinson's disease (PD). Up to date, the role of LRRK2 in PD onset and progression remains largely unknown. However, experimental evidence highlights a critical role of LRRK2 in the control of vesicle trafficking, likely by Rab phosphorylation, that in turn may regulate different aspects of neuronal physiology. Here we show that LRRK2 interacts with Sec8, one of eight subunits of the exocyst complex. The exocyst complex is an evolutionarily conserved multisubunit protein complex mainly involved in tethering secretory vesicles to the plasma membrane and implicated in the regulation of multiple biological processes modulated by vesicle trafficking. Interestingly, Rabs and exocyst complex belong to the same protein network. Our experimental evidence indicates that LRRK2 kinase activity or the presence of the LRRK2 kinase domain regulate the assembly of exocyst subunits and that the over-expression of Sec8 significantly rescues the LRRK2 G2019S mutant pathological effect. Our findings strongly suggest an interesting molecular mechanism by which LRRK2 could modulate vesicle trafficking and may have important implications to decode the complex role that LRRK2 plays in neuronal physiology.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Vesicular Transport Proteins/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice, Knockout , PC12 Cells , Protein Binding , Rats
8.
Cell Death Dis ; 11(5): 369, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409664

ABSTRACT

TDP-43 pathology is a disease hallmark that characterizes both amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD-TDP). TDP-43 undergoes several posttranslational modifications that can change its biological activities and its aggregative propensity, which is a common hallmark of different neurodegenerative conditions. New evidence is provided by the current study pointing at TDP-43 acetylation in ALS cellular models. Using both in vitro and in vivo approaches, we demonstrate that TDP-43 interacts with histone deacetylase 1 (HDAC1) via RRM1 and RRM2 domains, that are known to contain the two major TDP-43 acetylation sites, K142 and K192. Moreover, we show that TDP-43 is a direct transcriptional activator of CHOP promoter and this activity is regulated by acetylation. Finally and most importantly, we observe both in cell culture and in Drosophila that a HDCA1 reduced level (genomic inactivation or siRNA) or treatment with pan-HDAC inhibitors exert a protective role against WT or pathological mutant TDP-43 toxicity, suggesting TDP-43 acetylation as a new potential therapeutic target. HDAC inhibition efficacy in neurodegeneration has long been debated, but future investigations are warranted in this area. Selection of more specific HDAC inhibitors is still a promising option for neuronal protection especially as HDAC1 appears as a downstream target of both TDP- 43 and FUS, another ALS-related gene.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cell Death/drug effects , DNA-Binding Proteins/pharmacology , Histone Deacetylase 1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Death/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Histone Deacetylase 1/genetics , Humans , Inclusion Bodies/metabolism , Mice , Mutation/genetics
9.
J Cell Mol Med ; 23(12): 8505-8510, 2019 12.
Article in English | MEDLINE | ID: mdl-31560168

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD). The LRRK2 physiological and pathological function is still debated. However, different experimental evidence based on LRRK2 cellular localization and LRRK2 protein interactors suggests that LRRK2 may be part and regulate a protein network modulating vesicle dynamics/trafficking. Interestingly, the synaptic vesicle protein SV2A is part of this protein complex. Importantly, SV2A is the binding site of the levetiracetam (LEV), a compound largely used in human therapy for epilepsy treatment. The binding of LEV to SV2A reduces the neuronal firing by the modulation of vesicle trafficking although by an unclear molecular mechanism. In this short communication, we have analysed the interaction between the LRRK2 and SV2A pathways by LEV treatment. Interestingly, LEV significantly counteracts the effect of LRRK2 G2019S pathological mutant expression in three different cellular experimental models. Our data strongly suggest that LEV treatment may have a neuroprotective effect on LRRK2 pathological mutant toxicity and that LEV repositioning could be a viable compound for PD treatment.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Levetiracetam/pharmacology , Mutation , Neurons/drug effects , Synaptic Vesicles/metabolism , Animals , Anticonvulsants/pharmacology , Cell Line, Tumor , Cells, Cultured , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Neurites/physiology , Neurons/metabolism , Neurons/physiology , PC12 Cells , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Binding , Rats , Signal Transduction/drug effects
10.
Neuroscience ; 390: 1-11, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30134203

ABSTRACT

Neurodegenerative disorders, including Amyotrophic Lateral Sclerosis (ALS), have been associated to alterations in chromatin structure resulting in long-lasting changes in gene expression. ALS is predominantly a sporadic disease and environmental triggers may be involved in its onset. In this respect, alterations in the epigenome can provide the key to transform the genetic information into phenotype. In this paper, we demonstrate that two modifications associated with transcriptional activation, namely dimethylation of lysine 4 on H3 tail (H3K4me2) and phospho-acetylation of serine 10 and lysine 14 on H3 tail (H3K14ac-S10ph), and two modifications associated to transcriptional repression, namely trimethylation of lysine 9 on H3 tail (H3K9me3) and DNA methylation are selectively altered in cellular and animal model of ALS. These results reinforce the idea that epigenetic therapy may represent a potential and attractive approach for ALS treatment.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Epigenesis, Genetic , Protein Processing, Post-Translational , Animals , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/metabolism , HEK293 Cells , Histones/metabolism , Humans , Mice, Transgenic , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase-1/metabolism
11.
ACS Med Chem Lett ; 8(9): 941-946, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28947941

ABSTRACT

We report the synthesis, biological evaluation, and structural study of a series of substituted heteroaryl-pyrazole carboxylic acid derivatives. These compounds have been developed as inhibitors of specific isoforms of carbonic anhydrase (CA), with potential as prototypes of a new class of chemotherapeutics. Both X-ray crystallography and computational modeling provide insights into the CA inhibition mechanism. Results indicate that this chemotype produces an indirect interference with the zinc ion, thus behaving differently from other related nonclassical inhibitors. Among the tested compounds, 2c with Ki = 0.21 µM toward hCA XII demonstrated significant antiproliferative activity against hypoxic tumor cell lines. Taken together, the results thus provide the basis of structural determinants for the development of novel anticancer agents.

12.
PLoS One ; 12(6): e0179082, 2017.
Article in English | MEDLINE | ID: mdl-28582422

ABSTRACT

Mutations in LRRK2 play a critical role in both familial and sporadic Parkinson's disease (PD). Up to date, the role of LRRK2 in PD onset and progression remains largely unknown. However, experimental evidence highlights a critical role of LRRK2 in the control of vesicle trafficking that in turn may regulate different aspects of neuronal physiology. We have analyzed the role of LRRK2 in regulating dopamine receptor D1 (DRD1) and D2 (DRD2) trafficking. DRD1 and DRD2 are the most abundant dopamine receptors in the brain. They differ in structural, pharmacological and biochemical properties, as well as in localization and internalization mechanisms. Our results indicate that disease-associated mutant G2019S LRRK2 impairs DRD1 internalization, leading to an alteration in signal transduction. Moreover, the mutant forms of LRRK2 affect receptor turnover by decreasing the rate of DRD2 trafficking from the Golgi complex to the cell membrane. Collectively, our findings are consistent with the conclusion that LRRK2 influences the motility of neuronal vesicles and the neuronal receptor trafficking. These findings have important implications for the complex role that LRRK2 plays in neuronal physiology and the possible pathological mechanisms that may lead to neuronal death in PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Neurons/metabolism , Parkinson Disease/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Golgi Apparatus/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Transport , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction
13.
Rev Neurosci ; 28(2): 133-144, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28030361

ABSTRACT

Redox processes are key events in the degenerative cascade of many adult-onset neurodegenerative diseases (NDs), but the biological relevance of a single redox change is often dependent on the redox couple involved and on its subcellular origin. The biosensors based on engineered fluorescent proteins (redox-sensitive GFP [roGFP]) offer a unique opportunity to monitor redox changes in both physiological and pathological contexts in living animals and plants. Here, we review the use of roGFPs to monitor oxidative stress in different three adult-onset NDs: Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Despite the many differences spanning from incidence to onset, the hypotheses on biological processes underlying both sporadic and familiar ND forms in humans outline a model in which noncompeting mechanisms are likely to converge in various unsuccessful patterns to mediate the selective degeneration of a specific neuronal population. roGFPs, targeted to different cell compartments, are successfully used as specific markers of cell toxicity, induced by expression of causative genes linked to a determined ND. We also report the use of roGFP to monitor oxidative stress induced by the expression of the ALS-causative gene SOD1.


Subject(s)
Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Parkinson Disease/metabolism , Alzheimer Disease/diagnosis , Amyotrophic Lateral Sclerosis/diagnosis , Animals , Brain/metabolism , Brain/physiopathology , Humans , Parkinson Disease/diagnosis
14.
Int J Nanomedicine ; 9: 4935-51, 2014.
Article in English | MEDLINE | ID: mdl-25364251

ABSTRACT

BACKGROUND: Gold nanoparticles (GNPs) are likely to provide an attractive platform for combining a variety of biophysicochemical properties into a unified nanodevice with great therapeutic potential. In this study we investigated the capabilities of three different natural polyphenols, epigallocatechin-3-gallate (EGCG), resveratrol (RSV), and fisetin (FS), to allow synergistic chemical reduction of gold salts to GNPs and stabilization in a single-step green process. Moreover, antioxidant properties of the nanosystems, as well as preliminary antiproliferative activity and apoptotic process investigation of model EGCG-GNPs on stable clones of neuroblastoma SH-SY5Y cells expressing CFP-DEVD-YFP reporter, were examined. METHODS: The GNPs were characterized by physicochemical techniques, polyphenol content, and in vitro stability. The antioxidant activity of the GNPs was also determined by 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) cation (ABTS) radical-scavenging assays. Stable clones of neuronal SH-SY5Y-CFP-DEVD-YFP were generated and characterized, and cell viability after treatment with EGCG-GNPs was assessed after 72 hours through a 3(4,5-dimethylthiazol-2yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Activation of the apoptotic pathways was also investigated by Western blot analysis. RESULTS: With a diameter in the size range of 10-25 nm, the obtained nanoparticles (NPs) were found to contain 2.71%, 3.23%, and 5.47% of EGCG, RSV, and FS, respectively. Nanoprototypes exhibited remarkable in vitro stability in various media, suggesting that NP surface coating with phytochemicals prevents aggregation in different simulated physiological conditions. The scavenging activities for DPPH and ABTS were highly correlated with EGCG, RSV, and FS content. Moreover, high correlation coefficients between the ABTS and DPPH values were found for the prepared nanosystems. EGCG-GNPs induce a dose-dependent reduction on SH-SY5Y-CFP-DEVD-YFP cell viability that is likely to involve the activation of the apoptotic pathways, similarly to free EGCG, as suggested by the processing of the CFP-DEVD-YFP reporter. CONCLUSION: These results prompted us to propose the ecofriendly synthesized EGCG-, RSV-, and FS-based nanogold conjugates as suitable carriers for bioactive polyphenols to be used for the treatment of disorders associated with oxidative stress, including neurodegenerative disorders, cardiovascular disease, and cancer.


Subject(s)
Antioxidants/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Polyphenols/chemistry , Antioxidants/analysis , Antioxidants/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Catechin/analysis , Catechin/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Stability , Flavonoids/analysis , Flavonoids/chemistry , Flavonols , Green Chemistry Technology , Humans , Metal Nanoparticles/toxicity , Polyphenols/analysis , Polyphenols/pharmacology , Resveratrol , Stilbenes/analysis , Stilbenes/chemistry
15.
J Neurochem ; 131(2): 239-50, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24947832

ABSTRACT

Genetic studies show that LRRK2, and not its closest paralogue LRRK1, is linked to Parkinson's disease. To gain insight into the molecular and cellular basis of this discrepancy, we searched for LRRK1- and LRRK2-specific cellular processes by identifying their distinct interacting proteins. A protein microarray-based interaction screen was performed with recombinant 3xFlag-LRRK1 and 3xFlag-LRRK2 and, in parallel, co-immunoprecipitation followed by mass spectrometry was performed from SH-SY5Y neuroblastoma cell lines stably expressing 3xFlag-LRRK1 or 3xFlag-LRRK2. We identified a set of LRRK1- and LRRK2-specific as well as common interactors. One of our most prominent findings was that both screens pointed to epidermal growth factor receptor (EGF-R) as a LRRK1-specific interactor, while 14-3-3 proteins were LRRK2-specific. This is consistent with phosphosite mapping of LRRK1, revealing phosphosites outside of 14-3-3 consensus binding motifs. To assess the functional relevance of these interactions, SH-SY5Y-LRRK1 and -LRRK2 cell lines were treated with LRRK2 kinase inhibitors that disrupt 14-3-3 binding, or with EGF, an EGF-R agonist. Redistribution of LRRK2, not LRRK1, from diffuse cytoplasmic to filamentous aggregates was observed after inhibitor treatment. Similarly, EGF induced translocation of LRRK1, but not of LRRK2, to endosomes. Our study confirms that LRRK1 and LRRK2 can carry out distinct functions by interacting with different cellular proteins. LRRK1 and LRRK2 (leucine-rich repeat kinase) interaction partners were identified by two different protein-protein interaction screens. These confirmed epidermal growth factor receptor (EGR-R) as a LRRK1-specific interactor, while 14-3-3 proteins were LRRK2-specific. Functional analysis of these interactions and the pathways they mediate shows that LRRK1 and LRRK2 signaling do not intersect, reflective of the differential role of both LRRKs in Parkinson's disease.


Subject(s)
Protein Interaction Domains and Motifs/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2
16.
PLoS One ; 8(10): e77198, 2013.
Article in English | MEDLINE | ID: mdl-24167564

ABSTRACT

The leucine-rich repeat kinase 2 (LRRK2) gene was found to play a role in the pathogenesis of both familial and sporadic Parkinson's disease (PD). LRRK2 encodes a large multi-domain protein that is expressed in different tissues. To date, the physiological and pathological functions of LRRK2 are not clearly defined. In this study we have explored the role of LRRK2 in controlling vesicle trafficking in different cellular or animal models and using various readouts. In neuronal cells, the presence of LRRK2(G2019S) pathological mutant determines increased extracellular dopamine levels either under basal conditions or upon nicotine stimulation. Moreover, mutant LRRK2 affects the levels of dopamine receptor D1 on the membrane surface in neuronal cells or animal models. Ultrastructural analysis of PC12-derived cells expressing mutant LRRK2(G2019S) shows an altered intracellular vesicle distribution. Taken together, our results point to the key role of LRRK2 to control vesicle trafficking in neuronal cells.


Subject(s)
Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Dopamine D1/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , Dopamine/genetics , Dopamine/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mutation, Missense , Neurons/pathology , PC12 Cells , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics , Protein Transport/genetics , Rats , Receptors, Dopamine D1/genetics
17.
Biochem Soc Trans ; 40(5): 1117-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22988875

ABSTRACT

Mutations in LRRK2 (leucine-rich repeat kinase 2) (also known as PARK8 or dardarin) are responsible for the autosomal-dominant form of PD (Parkinson's disease). LRRK2 mutations were found in approximately 3-5% of familial and 1-3% of sporadic PD cases with the highest prevalence (up to 40%) in North Africans and Ashkenazi Jews. To date, mutations in LRRK2 are a major genetic risk factor for familial and sporadic PD. Despite the fact that 8 years have passed from the establishment of the first link between PD and dardarin in 2004, the pathophysiological role of LRRK2 in PD onset and progression is far from clearly defined. Also the generation of different LRRK2 transgenic or knockout animals has not provided new hints on the function of LRRK2 in the brain. The present paper reviews recent evidence regarding a potential role of LRRK2 in the regulation of membrane trafficking from vesicle generation to the movement along cytoskeleton and finally to vesicle fusion with cell membrane.


Subject(s)
Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Membrane/enzymology , Cytoplasmic Vesicles/enzymology , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation , Parkinson Disease/enzymology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/genetics
18.
J Neurosci ; 32(26): 9023-34, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22745501

ABSTRACT

Dysfunctions of dopaminergic homeostasis leading to either low or high dopamine (DA) levels are causally linked to Parkinson's disease, schizophrenia, and addiction. Major sites of DA synthesis are the mesencephalic neurons originating in the substantia nigra and ventral tegmental area; these structures send major projections to the dorsal striatum (DSt) and nucleus accumbens (NAcc), respectively. DA finely tunes its own synthesis and release by activating DA D2 receptors (D2R). To date, this critical D2R-dependent function was thought to be solely due to activation of D2Rs on dopaminergic neurons (D2 autoreceptors); instead, using site-specific D2R knock-out mice, we uncover that D2 heteroreceptors located on non-DAergic medium spiny neurons participate in the control of DA levels. This D2 heteroreceptor-mediated mechanism is more efficient in the DSt than in NAcc, indicating that D2R signaling differentially regulates mesolimbic- versus nigrostriatal-mediated functions. This study reveals previously unappreciated control of DA signaling, shedding new light on region-specific regulation of DA-mediated effects.


Subject(s)
Dopamine/metabolism , Neurons/cytology , Neurons/physiology , Presynaptic Terminals/metabolism , Receptors, Dopamine D2/metabolism , Synapses/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Biophysics , Chromatography, High Pressure Liquid/methods , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation/methods , Electrochemical Techniques , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Homovanillic Acid/metabolism , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Mutation/genetics , Neurons/drug effects , Patch-Clamp Techniques , Phosphorylation/drug effects , Phosphorylation/genetics , Presynaptic Terminals/drug effects , Quinpirole/pharmacology , RNA, Messenger/metabolism , Reaction Time/genetics , Receptors, Dopamine D2/genetics , Substantia Nigra/cytology , Substantia Nigra/drug effects , Synapses/drug effects , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects
19.
Neurobiol Dis ; 43(3): 642-50, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21624464

ABSTRACT

Expression of mutant SOD1 typical of familial amyotrophic lateral sclerosis (ALS) induces the expression of Bcl2-A1, a member of the Bcl2 family of proteins, specifically in motor neurons of transgenic mice. In this work, we have used immortalized motor neurons (NSC-34) and transgenic mice expressing mutant SOD1 to unravel the molecular mechanisms and the biological meaning of this up-regulation. We report that up-regulation of Bcl2-A1 by mutant SOD1 is mediated by activation of the redox sensitive transcription factor AP1 and that Bcl2-A1 interacts with pro-caspase-3 via its C-terminal helix α9. Furthermore, Bcl2-A1 inhibits pro-caspase-3 activation in immortalized motor neurons expressing mutant SOD1 and thus induction of Bcl2-A1 in ALS mice represents a pro-survival strategy aimed at counteracting the toxic effects of mutant SOD1. These data provide significant new insights on how molecular signaling, driven by expression of the ALS-causative gene SOD1, affects regulation of apoptosis in motor neurons and thus may have implications for ALS therapy, where prevention of motor neuronal cell death is one of the major aims.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Apoptosis/genetics , Caspase 3/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/enzymology , Animals , Caspase 3/metabolism , Caspase Inhibitors , Cell Line, Transformed , Cell Survival/genetics , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens , Motor Neurons/enzymology , Motor Neurons/metabolism , Motor Neurons/pathology , Oxidation-Reduction , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation/genetics
20.
Front Biosci (Elite Ed) ; 3(3): 1034-41, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622112

ABSTRACT

The health-promoting effects of fruit and vegetable consumption are thought to be due to phytochemicals contained in fresh plant material. Whether processed plant foods provide the same benefits as unprocessed ones is an open question. Melanoidins from heat-processed plums (prunes) were isolated and their presence confirmed by hydroxymethylfurfural content and browning index. Oxidative-induced endothelial cell (EC) damage is the trigger for the development of cardiovascular diseases (CVD); therefore the potential protective effect of prune melanoidins on hydrogen peroxide-induced oxidative cell damage was investigated on human endothelial ECV304 cells. Cytoplasmic and mitochondrial redox status was assessed by using the novel, redox-sensitive, ratiometric fluorescent protein sensor (roGFP), while mitochondrial membrane potential (MMP) was investigated with the fluorescent dye, JC-1. Treatment of ECV304 cells with hydrogen peroxide dose-dependently induced both mitochondrial and cytoplasmic oxidation, in addition to MMP dissipation, with ensuing cell death. Pretreatment of ECV304 with prune melanoidins, significantly counteracted and ultimately abolished hydrogen peroxide elicited phenomena, clearly indicating that these polymers protect human EC against oxidative stress.


Subject(s)
Cell Death/drug effects , Endothelium, Vascular/drug effects , Oxidative Stress/drug effects , Polymers/pharmacology , Prunus/chemistry , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology
SELECTION OF CITATIONS
SEARCH DETAIL