32 results on '"Sara Saez-Atienzar"'
Search Results
2. Reply to Zhu et al.: Implications of
- Author
-
Ruth, Chia, Sara, Saez-Atienzar, Daniel B, Drachman, and Bryan J, Traynor
- Subjects
Receptor, ErbB-2 ,Myasthenia Gravis ,Humans ,Receptors, Nicotinic ,Thymectomy - Published
- 2022
3. Large-scale pathway specific polygenic risk and transcriptomic community network analysis identifies novel functional pathways in Parkinson disease
- Author
-
Anthony R. Soltis, Ziv Gan-Or, Clifton L. Dalgard, Debra Ehrlich, Leonard H, Sara Saez-Atienzar, Cornelis Blauwendraat, Ali Torkamani, J. R. Gibbs, Sonja W. Scholz, Bryan J. Traynor, Clemens R. Scherzer, Jonggeol Jeff Kim, Jinhui Ding, Mark R. Cookson, Juan A. Botía, Matt Bookman, Andrew B. Singleton, Mike A. Nalls, Sara Bandres-Ciga, Monica Diez-Fairen, Hirotaka Iwaki, Lasse Pihlstrøm, Alastair J. Noyce, Dena G. Hernandez, Mina Ryten, Mary B. Makarious, and Faraz Faghri
- Subjects
Original Paper ,Polygenic risk ,Transcriptome community maps ,Context (language use) ,Computational biology ,Disease ,Quantitative trait locus ,Biology ,Chromatin remodeling ,Pathology and Forensic Medicine ,Parkinson disease ,Transcriptome ,Cellular and Molecular Neuroscience ,Mendelian randomization ,Neurology (clinical) ,Allele ,Signal transduction ,Gene - Abstract
Polygenic inheritance plays a central role in Parkinson disease (PD). A priority in elucidating PD etiology lies in defining the biological basis of genetic risk. Unraveling how risk leads to disruption will yield disease-modifying therapeutic targets that may be effective. Here, we utilized a high-throughput and hypothesis-free approach to determine biological processes underlying PD using the largest currently available cohorts of genetic and gene expression data from International Parkinson’s Disease Genetics Consortium (IPDGC) and the Accelerating Medicines Partnership-Parkinson’s disease initiative (AMP-PD), among other sources. We applied large-scale gene-set specific polygenic risk score (PRS) analyses to assess the role of common variation on PD risk focusing on publicly annotated gene sets representative of curated pathways. We nominated specific molecular sub-processes underlying protein misfolding and aggregation, post-translational protein modification, immune response, membrane and intracellular trafficking, lipid and vitamin metabolism, synaptic transmission, endosomal–lysosomal dysfunction, chromatin remodeling and apoptosis mediated by caspases among the main contributors to PD etiology. We assessed the impact of rare variation on PD risk in an independent cohort of whole-genome sequencing data and found evidence for a burden of rare damaging alleles in a range of processes, including neuronal transmission-related pathways and immune response. We explored enrichment linked to expression cell specificity patterns using single-cell gene expression data and demonstrated a significant risk pattern for dopaminergic neurons, serotonergic neurons, hypothalamic GABAergic neurons, and neural progenitors. Subsequently, we created a novel way of building de novo pathways by constructing a network expression community map using transcriptomic data derived from the blood of PD patients, which revealed functional enrichment in inflammatory signaling pathways, cell death machinery related processes, and dysregulation of mitochondrial homeostasis. Our analyses highlight several specific promising pathways and genes for functional prioritization and provide a cellular context in which such work should be done. Electronic supplementary material The online version of this article (10.1007/s00401-020-02181-3) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
4. Differential Methylation Analysis in Neuropathologically Confirmed Dementia with Lewy Bodies
- Author
-
Paolo Reho, Sultana Solaiman, Zalak Shah, Sara Saez-Atienzar, Ruth Chia, Karri Kaivola, Bryan J. Traynor, Bension S. Tilley, Steve M. Gentleman, Angela K. Hodges, Dag Aarsland, Edwin S. Monuki, Kathy L. Newell, Randall L. Woltjer, Marilyn S. Albert, Ted M. Dawson, Liana S. Rosenthal, Juan C. Troncoso, Olga Pletnikova, Geidy E. Serrano, Thomas G. Beach, Hariharan P. Easwaran, and Sonja Scholz
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
5. Genetic analysis of amyotrophic lateral sclerosis identifies contributing pathways and cell types
- Author
-
Ramita Dewan, Mike A. Nalls, Sarah Ahmed, Sara Bandres-Ciga, Bryan J. Traynor, Ruth Chia, Yevgeniya Abramzon, Italsgen, Mina Ryten, Sara Saez-Atienzar, Mark R. Cookson, Regina H. Reynolds, John Landers, Shing Wan Choi, Jonggeol J. Kim, Rebekah G. Langston, and Adriano Chiò
- Subjects
Cell type ,Disease ,Biology ,Polymorphism, Single Nucleotide ,Genetic analysis ,Biological pathway ,03 medical and health sciences ,0302 clinical medicine ,Neuron projection morphogenesis ,Mendelian randomization ,Genetics ,medicine ,Humans ,Genetic Testing ,Amyotrophic lateral sclerosis ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Amyotrophic Lateral Sclerosis ,SciAdv r-articles ,medicine.disease ,Signal transduction ,Neuroscience ,030217 neurology & neurosurgery ,Research Article ,Genome-Wide Association Study - Abstract
Massive genetic analysis identifies critical pathways and cell types involved in pathogenesis of amyotrophic lateral sclerosis., Despite the considerable progress in unraveling the genetic causes of amyotrophic lateral sclerosis (ALS), we do not fully understand the molecular mechanisms underlying the disease. We analyzed genome-wide data involving 78,500 individuals using a polygenic risk score approach to identify the biological pathways and cell types involved in ALS. This data-driven approach identified multiple aspects of the biology underlying the disease that resolved into broader themes, namely, neuron projection morphogenesis, membrane trafficking, and signal transduction mediated by ribonucleotides. We also found that genomic risk in ALS maps consistently to GABAergic interneurons and oligodendrocytes, as confirmed in human single-nucleus RNA-seq data. Using two-sample Mendelian randomization, we nominated six differentially expressed genes (ATG16L2, ACSL5, MAP1LC3A, MAPKAPK3, PLXNB2, and SCFD1) within the significant pathways as relevant to ALS. We conclude that the disparate genetic etiologies of this fatal neurological disease converge on a smaller number of final common pathways and cell types.
- Published
- 2021
- Full Text
- View/download PDF
6. LRRK2 mediates tubulation and vesicle sorting from lysosomes
- Author
-
Alexandra Beilina, Ravindran Kumaran, Christopher K. E. Bleck, Natalie Landeck, Yan Li, Jillian H. Kluss, Sara Saez-Atienzar, Eric Lindberg, Adamantios Mamais, Mark R. Cookson, Luis Bonet-Ponce, and Chad D. Williamson
- Subjects
Proteomics ,Lysosomal membrane ,Diseases and Disorders ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,03 medical and health sciences ,0302 clinical medicine ,Lysosome ,medicine ,Phosphorylation ,Research Articles ,030304 developmental biology ,LRRK2 Gene ,0303 health sciences ,Multidisciplinary ,Chemistry ,Vesicle ,SciAdv r-articles ,Signal transducing adaptor protein ,Cell Biology ,LRRK2 ,nervous system diseases ,Cell biology ,Protein Transport ,medicine.anatomical_structure ,Mutation ,Lysosomes ,030217 neurology & neurosurgery ,Research Article - Abstract
The Parkinson disease kinase LRRK2 translocates to the lysosomal membrane triggering lysosomal sorting through JIP4., Genetic variation around the LRRK2 gene affects risk of both familial and sporadic Parkinson’s disease (PD). However, the biological functions of LRRK2 remain incompletely understood. Here, we report that LRRK2 is recruited to lysosomes after exposure of cells to the lysosome membrane–rupturing agent LLOME. Using an unbiased proteomic screen, we identified the motor adaptor protein JIP4 as an LRRK2 partner at the lysosomal membrane. LRRK2 can recruit JIP4 to lysosomes in a kinase-dependent manner via the phosphorylation of RAB35 and RAB10. Using super-resolution live-cell imaging microscopy and FIB-SEM, we demonstrate that JIP4 promotes the formation of LAMP1-negative tubules that release membranous content from lysosomes. Thus, we describe a new process orchestrated by LRRK2, which we name LYTL (LYsosomal Tubulation/sorting driven by LRRK2), by which lysosomal tubulation is used to release vesicles from lysosomes. Given the central role of the lysosome in PD, LYTL is likely to be disease relevant.
- Published
- 2020
- Full Text
- View/download PDF
7. Author Correction: Cellular senescence and Alzheimer disease: the egg and the chicken scenario
- Author
-
Sara Saez-Atienzar and Eliezer Masliah
- Subjects
General Neuroscience ,medicine ,MEDLINE ,Cellular senescence ,Alzheimer's disease ,Biology ,Bioinformatics ,medicine.disease - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
8. Genetic analysis of amyotrophic lateral sclerosis identifies contributing pathways and cell types
- Author
-
Mina Ryten, Italsgen, Yevgeniya Abramzon, Sara Saez-Atienzar, Mark R. Cookson, Ramita Dewan, Mike A. Nalls, Sarah Ahmed, Rebekah G. Langston, Regina H. Reynolds, Adriano Chiò, Jonggeol J. Kim, Shing Wan Choi, Sara Bandres-Ciga, John Landers, Bryan J. Traynor, and Ruth Chia
- Subjects
Biological pathway ,Cell type ,Neuron projection morphogenesis ,Mendelian randomization ,medicine ,Disease ,Signal transduction ,Biology ,Amyotrophic lateral sclerosis ,medicine.disease ,Neuroscience ,Genetic analysis - Abstract
Despite the considerable progress in unraveling the genetic causes of amyotrophic lateral sclerosis (ALS), we do not fully understand the molecular mechanisms underlying the disease. We analyzed genome-wide data involving 78,500 individuals using a polygenic risk score approach to identify the biological pathways and cell types involved in ALS. This data-driven approach identified multiple aspects of the biology underlying the disease that resolved into broader themes, namely neuron projection morphogenesis, membrane trafficking, and signal transduction mediated by ribonucleotides. We also found that genomic risk in ALS maps consistently to GABAergic cortical interneurons and oligodendrocytes, as confirmed in human single-nucleus RNA-seq data. Using two-sample Mendelian randomization, we nominated five differentially expressed genes (ATG16L2, ACSL5, MAP1LC3A, PLXNB2, and SCFD1) within the significant pathways as relevant to ALS. We conclude that the disparate genetic etiologies of this fatal neurological disease converge on a smaller number of final common pathways and cell types.
- Published
- 2020
- Full Text
- View/download PDF
9. Generation of two induced pluripotent stem cell (iPSC) lines from an ALS patient with simultaneous mutations in KIF5A and MATR3 genes
- Author
-
Marissa Dominick, Ileana Lorenzini, Robert Bowser, Bryan J. Traynor, Sara Saez-Atienzar, David X. Medina, Ashley Boehringer, Rita Sattler, and Erik P. Pioro
- Subjects
0301 basic medicine ,Disease mechanisms ,Karyotype ,Cell Biology ,General Medicine ,Germ layer ,Biology ,medicine.disease ,Viral vector ,Cell biology ,03 medical and health sciences ,MATR3 Gene ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,medicine ,Amyotrophic lateral sclerosis ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Gene ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Fibroblasts from an amyotrophic lateral sclerosis patient with simultaneous mutations in the MATR3 gene and KIF5A gene were isolated and reprogrammed into induced pluripotent stem cells via a non-integrating Sendai viral vector. The generated iPSC clones demonstrated normal karyotype, expression of pluripotency markers, and the capacity to differentiate into three germ layers. The unique presence of two simultaneous mutations in ALS-associated genes represent a novel tool for the study of ALS disease mechanisms.
- Published
- 2020
10. Genome sequencing analysis identifies new loci associated with Lewy body dementia and provides insights into the complex genetic architecture
- Author
-
Pentti J. Tienari, James B. Leverenz, Nahid Tayebi, Gabriele Mora, Bradley F. Boeve, Laura Palmer, Steve M. Gentleman, Ellen Sidransky, Pau Pastor, Liana S. Rosenthal, G. Xiromerisiou, Sara Saez-Atienzar, Francesco Landi, Scott M. Kaiser, Qinwen Mao, Claire Troakes, Peter St George-Hyslop, Andrea Calvo, Suzanne Lesage, Mario Masellis, Randy Woltjer, Marilyn S. Albert, Thomas T. Warner, Lorraine N. Clark, Gregory Klein, Charles Duyckaerts, Seth Love, Ed Monuki, Lawrence S. Honig, Kelley Faber, Dennis W. Dickson, Lucy Norcliffe-Kaufmann, Cornelis Blauwendraat, Ronald C. Kim, Kevin Morgan, Clifton L. Dalgard, Joshua T. Geiger, Ali Torkamani, Jinhui Ding, Juan Fortea, Eliezer Masliah, Ekaterina Rogaeva, Matthew H. Perkins, Clemens R. Scherzer, John Q. Trojanowski, Zbigniew K. Wszolek, Glenda M. Halliday, Jordi Clarimón, Sonja W. Scholz, Olaf Ansorge, Makayla K. Portley, Toshiko Tanaka, Mary B. Makarious, Safa Al-Sarraj, Giancarlo Logroscino, John D. Eicher, Neill R. Graff-Radford, Carmen Lage, Ziv Gan-Or, Francesca Brett, Alison Goate, Raffaele Ferrari, John C. Morris, J. Raphael Gibbs, Lynn M. Bekris, Jose-Alberto Palma, Angela K. Hodges, Regina H. Reynolds, Alexis Brice, Monica Diez-Fairen, Coralie Viollet, Patrick May, Minna Oinas, Erika Salvi, Vivianna M. Van Deerlin, Estrella Morenas-Rodríguez, Anni Moore, Zane Jaunmuktane, Eileen H. Bigio, Daniele Cusi, Douglas Galasko, Ruth Chia, Kathy L. Newell, Isabel Santana, Claudia Schulte, David Goldstein, Thomas Gasser, Owen A. Ross, Walter A. Kukull, Tatiana Foroud, Chad A. Caraway, David A. Bennett, Samreen Ahmed, Lilah M. Besser, Antonio Canosa, Daniel Alcolea, Yevgeniya Abramzon, Elisabet Londos, Laura Parkkinen, Sandra E. Black, Eric Topol, Marya S. Sabir, Olga Pletnikova, Grisel Lopez, Tanis J. Ferman, Johannes Attems, Matthew J. Barrett, Margaret E. Flanagan, Horacio Kaufmann, Stuart Pickering Brown, Jon Infante, Ryan C. Bohannan, Alberto Lleó, Eloy Rodríguez-Rodríguez, Huw R. Morris, Gianluca Floris, Ted M. Dawson, Maura Brunetti, Alan E. Renton, Andrew B. Singleton, Karen Marder, Alan J. Thomas, Pascual Sanchez-Juan, Adriano Chiò, Nigel J. Cairns, David J. Stone, Tammaryn Lashley, Mike A. Nalls, Bernardino Ghetti, Sara Bandres-Ciga, Zalak Shah, Ian G. McKeith, Susan M. Resnick, Julia Keith, Liisa Myllykangas, Diego Albani, Christopher M. Morris, Vikram Shakkottai, M. Ryten, Ronald L. Walton, Isabel González-Aramburu, Luigi Ferrucci, Bryan J. Traynor, Amanda B. Kuzma, Afina W. Lemstra, Thomas G. Beach, Juan C. Troncoso, Emil K. Gustavsson, Maurizio Grassano, John Hardy, Geidy E. Serrano, Rejko Krüger, Dag Aarsland, Bension S. Tilley, and Dena G. Hernandez
- Subjects
0303 health sciences ,Lewy body ,Disease ,Computational biology ,Biology ,medicine.disease ,DNA sequencing ,Genetic architecture ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Dementia ,Genetic risk ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology ,Genetic association - Abstract
The genetic basis of Lewy body dementia (LBD) is not well understood. Here, we performed whole-genome sequencing in large cohorts of LBD cases and neurologically healthy controls to study the genetic architecture of this understudied form of dementia and to generate a resource for the scientific community. Genome-wide association analysis identified five independent risk loci, whereas genome-wide gene-aggregation tests implicated mutations in the gene GBA. Genetic risk scores demonstrate that LBD shares risk profiles and pathways with Alzheimer’s and Parkinson’s disease, providing a deeper molecular understanding of the complex genetic architecture of this age-related neurodegenerative condition.
- Published
- 2020
- Full Text
- View/download PDF
11. Cellular senescence and Alzheimer disease: the egg and the chicken scenario
- Author
-
Eliezer Masliah and Sara Saez-Atienzar
- Subjects
0301 basic medicine ,Aging ,Microglia ,Mechanism (biology) ,General Neuroscience ,Neurodegeneration ,Context (language use) ,Disease ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Ageing ,Alzheimer Disease ,medicine ,Dementia ,Animals ,Humans ,Alzheimer's disease ,Neuroscience ,030217 neurology & neurosurgery ,Cellular Senescence - Abstract
Globally, 50 million people live with dementia, with Alzheimer disease (AD) being responsible for two-thirds of the total cases. As ageing is the main risk factor for dementia-related neurodegeneration, changes in the timing or nature of the cellular hallmarks of normal ageing might be key to understanding the events that convert normal ageing into neurodegeneration. Cellular senescence is a candidate mechanism that might be important for this conversion. Under persistent stress, as occurs in ageing, both postmitotic cells — including neurons — and proliferative cells — such as astrocytes and microglia, among others — can engender a state of chronic cellular senescence that is characterized by the secretion of pro-inflammatory molecules that promote the functional decline of tissues and organs. Ablation of senescent cells has been postulated as a promising therapeutic venue to target the ageing phenotype and, thus, prevent or mitigate ageing-related diseases. However, owing to a lack of evidence, it is not possible to label cellular senescence as a cause or a consequence of neurodegeneration. This Review examines cellular senescence in the context of ageing and AD, and discusses which of the processes — cellular senescence or AD — might come first. Ageing is the main risk factor for Alzheimer disease (AD), but the mechanisms connecting ageing to this disease remain incompletely understood. In this Review, Saez-Atienzar and Masliah examine whether cellular senescence may have a role in linking ageing and AD.
- Published
- 2020
12. Large-scale pathway-specific polygenic risk, transcriptomic community networks and functional inferences in Parkinson disease
- Author
-
Mary B. Makarious, Andrew B. Singleton, Lasse Pihlstrøm, Mark R. Cookson, Mina Ryten, Alastair J. Noyce, Debra Ehrlich, Ziv Gan-Or, Jonggeol Jeff Kim, Jinhui Ding, Bryan J. Traynor, Hirotaka Iwaki, Cornelis Blauwendraat, Juan A. Botía, Dena G. Hernandez, Matthew Bookman, Ali Torkamani, Monica Diez-Fairen, Sara Saez-Atienzar, Mike A. Nalls, Sonja W. Scholz, Hampton L. Leonard, Sara Bandres-Ciga, Clemens R. Scherzer, Faraz Faghri, and Raphael Gibbs
- Subjects
0303 health sciences ,Context (language use) ,Computational biology ,Disease ,Quantitative trait locus ,Biology ,3. Good health ,Biological pathway ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Cohort ,Polygenic risk score ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Polygenic inheritance plays a central role in Parkinson disease (PD). A priority in elucidating PD etiology lies in defining the biological basis of genetic risk. Unraveling how risk leads to disruption will yield disease-modifying therapeutic targets that may be effective. Here, we utilized a high-throughput and hypothesis-free approach to determine biological pathways underlying PD using the largest currently available cohorts of genetic data and gene expression data from International Parkinson’s Disease Genetics Consortium (IPDGC) and the Accelerating Medicines Partnership - Parkinson’s disease initiative (AMP-PD), among other sources. We placed these insights into a cellular context. We applied large-scale pathway-specific polygenic risk score (PRS) analyses to assess the role of common variation on PD risk in a cohort of 457,110 individuals by focusing on a compilation of 2,199 publicly annotated gene sets representative of curated pathways, of which we nominate 46 pathways associated with PD risk. We assessed the impact of rare variation on PD risk in an independent cohort of whole-genome sequencing data, including 4,331 individuals. We explored enrichment linked to expression cell specificity patterns using single-cell gene expression data and demonstrated a significant risk pattern for adult dopaminergic neurons, serotonergic neurons, and radial glia. Subsequently, we created a novel way of building de novo pathways by constructing a network expression community map using transcriptomic data derived from the blood of 1,612 PD patients, which revealed 54 connecting networks associated with PD. Our analyses highlight several promising pathways and genes for functional prioritization and provide a cellular context in which such work should be done.
- Published
- 2020
- Full Text
- View/download PDF
13. ALS-associated KIF5A mutations abolish autoinhibition resulting in a toxic gain of function
- Author
-
Desiree M. Baron, Adam R. Fenton, Sara Saez-Atienzar, Anthony Giampetruzzi, Aparna Sreeram, null Shankaracharya, Pamela J. Keagle, Victoria R. Doocy, Nathan J. Smith, Eric W. Danielson, Megan Andresano, Mary C. McCormack, Jaqueline Garcia, Valérie Bercier, Ludo Van Den Bosch, Jonathan R. Brent, Claudia Fallini, Bryan J. Traynor, Erika L.F. Holzbaur, and John E. Landers
- Subjects
Science & Technology ,MOTOR DOMAIN ,CARGO-BINDING ,Amyotrophic Lateral Sclerosis ,Kinesins ,PROTEIN ,STATISTICAL-MODEL ,Cell Biology ,AXONAL-TRANSPORT ,Axonal Transport ,Article ,AMYOTROPHIC-LATERAL-SCLEROSIS ,General Biochemistry, Genetics and Molecular Biology ,Gain of Function Mutation ,Mutation ,Humans ,NUCLEAR IMPORT ,LIGHT-CHAINS ,Life Sciences & Biomedicine ,PAGETS-DISEASE ,GENE-EXPRESSION - Abstract
Understanding the pathogenic mechanisms of disease mutations is critical to advancing treatments. ALS-associated mutations in the gene encoding the microtubule motor KIF5A result in skipping of exon 27 (KIF5AΔExon27) and the encoding of a protein with a novel 39 amino acid residue C-terminal sequence. Here, we report that expression of ALS-linked mutant KIF5A results in dysregulated motor activity, cellular mislocalization, altered axonal transport, and decreased neuronal survival. Single-molecule analysis revealed that the altered C terminus of mutant KIF5A results in a constitutively active state. Furthermore, mutant KIF5A possesses altered protein and RNA interactions and its expression results in altered gene expression/splicing. Taken together, our data support the hypothesis that causative ALS mutations result in a toxic gain of function in the intracellular motor KIF5A that disrupts intracellular trafficking and neuronal homeostasis. ispartof: CELL REPORTS vol:39 issue:1 ispartof: location:United States status: published
- Published
- 2022
- Full Text
- View/download PDF
14. Distinct Connectivity and Functionality of Aldehyde Dehydrogenase 1a1-Positive Nigrostriatal Dopaminergic Neurons in Motor Learning
- Author
-
Jie Dong, Huaibin Cai, Jia Yu, Sara Saez-Atienzar, Junbing Wu, Sarah L. Hawes, Zayd M. Khaliq, Vivian Chen, Zhenhua Liu, Weidong Le, Rebekah C. Evans, Da-Ting Lin, Lisa Chang, Lixin Sun, Jinhui Ding, Justin W. Kung, Nannan Yang, Bo Liang, Chengsong Xie, Ahsan Habib, and Beisha Tang
- Subjects
0301 basic medicine ,Parkinson's disease ,Dopamine ,Substantia nigra ,Mice, Transgenic ,Striatum ,Biology ,Medium spiny neuron ,General Biochemistry, Genetics and Molecular Biology ,Aldehyde Dehydrogenase 1 Family ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Learning ,lcsh:QH301-705.5 ,Motor skill ,Dopaminergic Neurons ,Dopaminergic ,Retinal Dehydrogenase ,medicine.disease ,Corpus Striatum ,Substantia Nigra ,030104 developmental biology ,nervous system ,lcsh:Biology (General) ,Motor learning ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Summary: Parkinson’s disease causes the most profound loss of the aldehyde dehydrogenase 1A1-positive (ALDH1A1+) nigrostriatal dopaminergic neuron (nDAN) subpopulation. The connectivity and functionality of ALDH1A1+ nDANs, however, remain poorly understood. Here, we show in rodent brains that ALDH1A1+ nDANs project predominantly to the rostral dorsal striatum, from which they also receive most monosynaptic inputs, indicating extensive reciprocal innervations with the striatal spiny projection neurons (SPNs). Functionally, genetic ablation of ALDH1A1+ nDANs causes severe impairments in motor skill learning, along with a reduction in high-speed walking. While dopamine replacement therapy accelerated walking speed, it failed to improve motor skill learning in ALDH1A1+ nDAN-ablated mice. Altogether, our study provides a comprehensive whole-brain connectivity map and reveals a key physiological function of ALDH1A1+ nDANs in motor skill acquisition, suggesting the motor learning processes require ALDH1A1+ nDANs to integrate diverse presynaptic inputs and supply dopamine with dynamic precision. : Wu et al. show that aldehyde dehydrogenase 1A1-positive nigrostriatal dopaminergic neurons, which display the most profound degeneration in Parkinson’s disease, are essential in the acquisition of skilled movements in rodent models. The proper synaptic inputs to these neurons are critical for providing the timely dopamine release required in the learning process. Keywords: aldehyde dehydrogenase 1a1, ALDH1A1, Parkinson’s disease, dopamine, dopaminergic neurons, substantia nigra, striatum, brain mapping, motor skill learning
- Published
- 2019
15. The Parkinson's Disease Protein LRRK2 Interacts with the GARP Complex to Promote Retrograde Transport to the trans-Golgi Network
- Author
-
David C. Gershlick, Morié Ishida, Dorien A. Roosen, Vlad Malkov, Luis Bonet-Ponce, Ravindran Kumaran, Matthew J. Fell, Alexandra Beilina, Alice Kaganovich, Kirsten Harvey, Jennifer J. Kordich, Juan S. Bonifacino, Sara Saez-Atienzar, Darren J. Moore, Laura Pellegrini, Adamantios Mamais, and Mark R. Cookson
- Subjects
0301 basic medicine ,Leucine-rich repeat kinase 2 ,Endosome ,Protein subunit ,Vesicular Transport Proteins ,Golgi Apparatus ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,symbols.namesake ,Mice ,neurodegenerative disease ,0302 clinical medicine ,medicine ,Animals ,Humans ,Kinase activity ,endosome ,lcsh:QH301-705.5 ,Kinase ,Chemistry ,membrane trafficking ,Neurodegeneration ,neurodegeneration ,Membrane Proteins ,GARP complex ,Parkinson Disease ,Golgi apparatus ,medicine.disease ,LRRK2 ,Cell biology ,nervous system diseases ,Protein Transport ,030104 developmental biology ,lcsh:Biology (General) ,lysosome ,symbols ,030217 neurology & neurosurgery ,Protein Binding ,trans-Golgi Network - Abstract
SUMMARY Mutations in Leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease (PD). However, the precise function of LRRK2 remains unclear. We report an interaction between LRRK2 and VPS52, a subunit of the Golgi-associated retrograde protein (GARP) complex that identifies a function of LRRK2 in regulating membrane fusion at the trans-Golgi network (TGN). At the TGN, LRRK2 further interacts with the Golgi SNAREs VAMP4 and Syntaxin-6 and acts as a scaffolding platform that stabilizes the GARP-SNAREs complex formation. Therefore, LRRK2 influences both retrograde and post-Golgi trafficking pathways in a manner dependent on its GTP binding and kinase activity. This action is exaggerated by mutations associated with Parkinson’s disease and can be blocked by kinase inhibitors. Disruption of GARP sensitizes dopamine neurons to mutant LRRK2 toxicity in C. elegans, showing that these pathways are interlinked in vivo and suggesting a link in PD., Graphical Abstract, In Brief Mutations in LRRK2 are linked to Parkinson’s disease. However, the cellular role of LRRK2 remains elusive. Using a combination of proteomics and imaging techniques, Beilina et al. describe a function of LRRK2 mediating endosome-TGN transport by scaffolding the GARP:Syntaxin-6 interaction, suggesting a connection between GARP and LRRK2 in Parkinson’s disease.
- Published
- 2018
16. On the mechanism underlying ethanol-induced mitochondrial dynamic disruption and autophagy response
- Author
-
Francisco J. Romero, Jorge M. Barcia, Maria F. Galindo, Carmen da Casa, Joaquín Jordán, Miguel Flores-Bellver, Javier Sancho-Pelluz, Luis Bonet-Ponce, and Sara Saez-Atienzar
- Subjects
Mitochondrial ROS ,Dynamins ,Programmed cell death ,Apoptosis ,Retinal Pigment Epithelium ,Biology ,Mitochondrial apoptosis-induced channel ,Mitochondrial Dynamics ,Autophagy-Related Protein 5 ,Cell Line ,GTP Phosphohydrolases ,Mice ,Mitophagy ,Animals ,Humans ,Drp-1 ,Molecular Biology ,bcl-2-Associated X Protein ,Ethanol ,Ribosomal Protein S6 Kinases ,Autophagy ,Membrane Proteins ,Fibroblasts ,Cell biology ,Mitochondria ,Mitochondrial permeability transition pore ,Oxidative stress ,Bax ,DNAJA3 ,Small Ubiquitin-Related Modifier Proteins ,Molecular Medicine ,Mitochondrial fission ,Beclin-1 ,Alcohol ,Apoptosis Regulatory Proteins ,Reactive Oxygen Species ,Microtubule-Associated Proteins ,Autophagy-Related Protein 12 - Abstract
We have explored the mechanisms underlying ethanol-induced mitochondrial dynamics disruption and mitophagy. Ethanol increases mitochondrial fission in a concentration-dependent manner through Drp1 mitochondrial translocation and OPA1 proteolytic cleavage. ARPE-19 (a human retinal pigment epithelial cell line) cells challenged with ethanol showed mitochondrial potential disruptions mediated by alterations in mitochondrial complex IV protein level and increases in mitochondrial reactive oxygen species production. In addition, ethanol activated the canonical autophagic pathway, as denoted by autophagosome formation and autophagy regulator elements including Beclin1, ATG5-ATG12 and P-S6 kinase. Likewise, autophagy inhibition dramatically increased mitochondrial fission and cell death, whereas autophagy stimulation rendered the opposite results, placing autophagy as a cytoprotective response aimed to remove damaged mitochondria. Interestingly, although ethanol induced mitochondrial Bax translocation, this episode was associated to cell death rather than mitochondrial fission or autophagy responses. Thus, Bax required 600mM ethanol to migrate to mitochondria, a concentration that resulted in cell death. Furthermore, mouse embryonic fibroblasts lacking this protein respond to ethanol by undergoing mitochondrial fission and autophagy but not cytotoxicity. Finally, by using the specific mitochondrial-targeted scavenger MitoQ, we revealed mitochondria as the main source of reactive oxygen species that trigger autophagy activation. These findings suggest that cells respond to ethanol activating mitochondrial fission machinery by Drp1 and OPA1 rather than bax, in a manner that stimulates cytoprotective autophagy through mitochondrial ROS.
- Published
- 2015
- Full Text
- View/download PDF
17. Automatic quantification of the subcellular localization of chimeric GFP protein supported by a two-level Naive Bayes classifier
- Author
-
Luis de la Ossa, Joaquín Jordán, Sara Saez-Atienzar, Jesus Martínez-Gómez, José M. Puerta, Maria F. Galindo, and Juan I. Alonso-Barba
- Subjects
Supervisor ,Computer science ,business.industry ,General Engineering ,Machine learning ,computer.software_genre ,Subcellular localization ,Cell counting ,Computer Science Applications ,Green fluorescent protein ,Naive Bayes classifier ,Artificial Intelligence ,Artificial intelligence ,business ,computer ,Classifier (UML) - Abstract
Automatic detection and counting of cells overexpressing GFP-chimeric proteins.Confocal microscopic images show two kinds of cells plus some objects which must be discarded.A trained specialist must distinguish and count manually the cells of each kind.The paper proposes a two level Naive Bayes classifier to carry out this task automatically.Results show that the proposed method is valid, and can avoid experts the manual quantification. Herein we report a method supported by a two-level Naive Bayes classifier to help and improve the automatic detection and counting of cells overexpressing GFP-chimeric proteins. This toll is frequently used as a reporter for the localization and the distribution pattern of a protein in a cell. This approximation requires, besides confocal microscopy, the participation of a qualified and blind counting supervisor to avoid subjective appreciations of the imaging interpretation of the data. Indeed, this counting required specific staff training, and the interpretation of the data is inevitably subjective. In order to avoid this, we have designed an automatic detection cell counting software. We have used as a model SH-SY5Y cells overexpressing GFP-Bax protein, after 6-hydroxydopamine addition. Our proposed method learns the counting criteria after a short training stage, and uses the resulting classifier to process new images and obtaining both the number of transfected cells and the proportion of these cells that present a translocated protein. The software achieves an accuracy over 97% when detecting transfected cells, and over 93% when detecting cells with GFP-Bax translocated. Besides the hours of qualified work that can be saved, the models learnt can be stored and reused (without training) so as to homogenize criteria among different researchers.
- Published
- 2015
- Full Text
- View/download PDF
18. Genetic risk factors in Parkinson's disease
- Author
-
Kimberley Billingsley, Andrew B. Singleton, Sara Bandres-Ciga, and Sara Saez-Atienzar
- Subjects
0301 basic medicine ,Histology ,Parkinson's disease ,Genome-wide association study ,Disease ,Computational biology ,Biology ,Disease pathogenesis ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Genetic Heterogeneity ,0302 clinical medicine ,Risk Factors ,medicine ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Genetic risk ,Genetic Association Studies ,Mechanism (biology) ,Parkinson Disease ,Cell Biology ,medicine.disease ,Human genetics ,Genetic architecture ,030104 developmental biology ,030217 neurology & neurosurgery - Abstract
Over the last two decades, we have witnessed a revolution in the field of Parkinson’s disease (PD) genetics. Great advances have been made in identifying many loci that confer a risk for PD, which has subsequently led to an improved understanding of the molecular pathways involved in disease pathogenesis. Despite this success, it is predicted that only a relatively small proportion of the phenotypic variability has been explained by genetics. Therefore, it is clear that common heritable components of disease are still to be identified. Dissecting the genetic architecture of PD constitutes a critical effort in identifying therapeutic targets and although such substantial progress has helped us to better understand disease mechanism, the route to PD disease-modifying drugs is a lengthy one. In this review, we give an overview of the known genetic risk factors in PD, focusing not on individual variants but the larger networks that have been implicated following comprehensive pathway analysis. We outline the challenges faced in the translation of risk loci to pathobiological relevance and illustrate the need for integrating big-data by noting success in recent work which adopts a broad-scale screening approach. Lastly, with PD genetics now progressing from identifying risk to predicting disease, we review how these models will likely have a significant impact in the future.
- Published
- 2017
19. Expression of the human TIMM23 and TIMM23B genes is regulated by the GABP transcription factor
- Author
-
Ignacio Ventura, José R. Blesa, Jesús A. Prieto-Ruiz, Rafael Alis, José Hernández-Yago, José Miguel Hernández-Andreu, Sara Saez-Atienzar, and Sandra García-Benlloch
- Subjects
0301 basic medicine ,Biophysics ,Biochemistry ,Mitochondrial Membrane Transport Proteins ,03 medical and health sciences ,Structural Biology ,Transcription (biology) ,Cell Line, Tumor ,Sequence Homology, Nucleic Acid ,Mitochondrial Precursor Protein Import Complex Proteins ,Genetics ,Humans ,Protein Isoforms ,Inner mitochondrial membrane ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Binding Sites ,030102 biochemistry & molecular biology ,Base Sequence ,RBPJ ,Chemistry ,Promoter ,GA-Binding Protein Transcription Factor ,Cell biology ,030104 developmental biology ,HEK293 Cells ,Mitochondrial biogenesis ,Gene Expression Regulation ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,Mutation ,RNA Interference ,Chromatin immunoprecipitation - Abstract
The TIM23 protein is a key component of the mitochondrial import machinery in yeast and mammals. TIM23 is the channel-forming subunit of the translocase of the inner mitochondrial membrane (TIM23) complex, which mediates preprotein translocation across the mitochondrial inner membrane. In this paper, we aimed to characterize the promoter region of the highly similar human TIM23 orthologs: TIMM23 and TIMM23B. Bioinformatic analysis revealed putative sites for the GA-binding protein (GABP) and the recombination signal binding protein for immunoglobulin kappa J (RBPJ) transcription factors in both promoters. Luciferase reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation experiments showed three functional sites for GABP and one functional site for RBPJ in both promoters. Moreover, silencing of GABPA, the gene encoding the DNA-binding subunit of the GABP transcription factor, resulted in reduced expression of TIMM23 and TIMM23B. Our results show an essential role of GABP in activating TIMM23 expression. More broadly, they suggest that physiological signals involved in activating mitochondrial biogenesis and oxidative function also enhance the transcription but not the protein level of TIMM23, which is essential for maintaining mitochondrial function and homeostasis.
- Published
- 2017
20. SATB1 is a Dopaminergic Neuron‐Specific Regulator of Cellular Senescence
- Author
-
Sara Saez-Atienzar and Alexandra Cancio‐Bello
- Subjects
business.industry ,Dopaminergic Neurons ,Regulator ,Cellular senescence ,Matrix Attachment Region Binding Proteins ,SATB1 ,Biology ,Article ,Text mining ,Neurology ,Disease Progression ,Humans ,Neurology (clinical) ,business ,Dopaminergic neuron ,Neuroscience ,Cellular Senescence - Published
- 2020
- Full Text
- View/download PDF
21. 3-Nitropropionic acid induces autophagy by forming mitochondrial permeability transition pores rather than activatiing the mitochondrial fission pathway
- Author
-
Maria F. Galindo, Joaquín Jordán, Sara Saez-Atienzar, and Maria E. Solesio
- Subjects
Pharmacology ,Mitochondrial membrane transport protein ,Mitochondrial permeability transition pore ,biology ,Cyclosporin a ,Autophagy ,biology.protein ,Mitochondrial fission ,ATP–ADP translocase ,Mitochondrion ,Mitochondrial apoptosis-induced channel ,Cell biology - Abstract
BACKGROUND AND PURPOSE Huntington's disease is a neurodegenerative process associated with mitochondrial alterations. Inhibitors of the electron–transport channel complex II, such as 3-nitropropionic acid (3NP), are used to study the molecular and cellular pathways involved in this disease. We studied the effect of 3NP on mitochondrial morphology and its involvement in macrophagy. EXPERIMENTAL APPROACH Pharmacological and biochemical methods were used to characterize the effects of 3NP on autophagy and mitochondrial morphology. SH-SY5Y cells were transfected with GFP-LC3, GFP-Drp1 or GFP-Bax to ascertain their role and intracellular localization after 3NP treatment using confocal microscopy. KEY RESULTS Untreated SH-SY5Y cells presented a long, tubular and filamentous net of mitochondria. After 3NP (5 mM) treatment, mitochondria became shorter and rounder. 3NP induced formation of mitochondrial permeability transition pores, both in cell cultures and in isolated liver mitochondria, and this process was inhibited by cyclosporin A. Participation of the mitochondrial fission pathway was excluded because 3NP did not induce translocation of the dynamin-related protein 1 (Drp1) to the mitochondria. The Drp1 inhibitor Mdivi-1 did not affect the observed changes in mitochondrial morphology. Finally, scavengers of reactive oxygen species failed to prevent mitochondrial alterations, while cyclosporin A, but not Mdivi-1, prevented the generation of ROS. CONCLUSIONS AND IMPLICATIONS There was a direct correlation between formation of mitochondrial permeability transition pores and autophagy induced by 3NP treatment. Activation of autophagy preceded the apoptotic process and was mediated, at least partly, by formation of reactive oxygen species and mitochondrial permeability transition pores. LINKED ARTICLE This article is commented on by Gonzalez-Polo et al., pp. 60–62 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2012.02203.x
- Published
- 2012
- Full Text
- View/download PDF
22. Rotenone Induces the Formation of 4-Hydroxynonenal Aggresomes. Role of ROS-Mediated Tubulin Hyperacetylation and Autophagic Flux Disruption
- Author
-
Eduardo Nava, Luis Bonet-Ponce, Jorge M. Barcia, Joaquín Jordán, Carmen da Casa, Francisco J. Romero, Javier Sancho-Pelluz, Sara Saez-Atienzar, Natalia Martínez-Gil, and Maria F. Galindo
- Subjects
0301 basic medicine ,Programmed cell death ,Neuroscience (miscellaneous) ,Mitochondrion ,Biology ,medicine.disease_cause ,Membrane Fusion ,Models, Biological ,Cell Line ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Protein Aggregates ,Tubulin ,Rotenone ,medicine ,Autophagy ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,Aldehydes ,Autophagosomes ,Acetylation ,Free radical scavenger ,Cell biology ,030104 developmental biology ,Aggresome ,Neurology ,chemistry ,Lysosomes ,Reactive Oxygen Species ,Oxidative stress - Abstract
Oxidative stress causes cellular damage by (i) altering protein stability, (ii) impairing organelle function, or (iii) triggering the formation of 4-HNE protein aggregates. The catabolic process known as autophagy is an antioxidant cellular response aimed to counteract these stressful conditions. Therefore, autophagy might act as a cytoprotective response by removing impaired organelles and aggregated proteins. In the present study, we sought to understand the role of autophagy in the clearance of 4-HNE protein aggregates in ARPE-19 cells under rotenone exposure. Rotenone induced an overproduction of reactive oxygen species (ROS), which led to an accumulation of 4-HNE inclusions, and an increase in the number of autophagosomes. The latter resulted from a disturbed autophagic flux rather than an activation of the autophagic synthesis pathway. In compliance with this, rotenone treatment induced an increase in LC3-II while upstream autophagy markers such as Beclin- 1, Vsp34 or Atg5-Atg12, were decreased. Rotenone reduced the autophagosome-to-lysosome fusion step by increasing tubulin acetylation levels through a ROS-mediated pathway. Proof of this is the finding that the free radical scavenger, N-acetylcysteine, restored autophagy flux and reduced rotenone-induced tubulin hyperacetylation. Indeed, this dysfunctional autophagic response exacerbates cell death triggered by rotenone, since 3-methyladenine, an autophagy inhibitor, reduced cell mortality, while rapamycin, an inductor of autophagy, caused opposite effects. In summary, we shed new light on the mechanisms involved in the autophagic responses disrupted by oxidative stress, which take place in neurodegenerative diseases such as Huntington or Parkinson diseases, and age-related macular degeneration.
- Published
- 2015
23. Bcl-xL-mediated antioxidant function abrogates the disruption of mitochondrial dynamics induced by LRRK2 inhibition
- Author
-
Eduardo Nava, Laura Perez-Dolz, Joaquín Jordán, Luis Bonet-Ponce, José R. Blesa, Sara Saez-Atienzar, Carmen da Casa, and Maria F. Galindo
- Subjects
0301 basic medicine ,Mitocondrial fission ,bcl-X Protein ,Bcl-xL ,medicine.disease_cause ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Mitochondrial Dynamics ,03 medical and health sciences ,Tubulin ,Cell Line, Tumor ,Acetylated tubulin ,medicine ,Autophagy ,Humans ,Parkinson ,Molecular Biology ,Dynamin ,biology ,Kinase ,Wild type ,LRRK2 ,Acetylation ,Parkinson Disease ,Molecular biology ,DRP-1 ,Cell biology ,Oxidative Stress ,030104 developmental biology ,Second messenger system ,biology.protein ,Molecular Medicine ,Oxidative stress - Abstract
We have used the human neuroblastoma cell line SH-SY5Y overexpressing Bcl-xL (SH-SY5Y/Bcl-xL) to clarify the effects of this mitochondrial protein on the control of mitochondrial dynamics and the autophagic processes which occur after the inhibition of leucine-rich repeat kinase 2 (LRRK2) with GSK2578215A. In wild type (SH-SY5Y/Neo) cells, GSK2578215A (1nM) caused a disruption of mitochondrial morphology and an imbalance in intracellular reactive oxygen species (ROS) as indicated by an increase in dichlorofluorescein fluorescence and 4-hydroxynonenal. However, SH-SY5Y/Bcl-xL cells under GSK2578215A treatment, unlike the wild type, preserved a high mitochondrial membrane potential and did not exhibit apoptotical chromatins. In contrast to wild type cells, in SH-SY5Y/Bcl-xL cells, GSK2578215A did not induce mitochondrial translocation of neither dynamin related protein-1 nor the proapoptotic protein, Bax. In SH-SY5Y/Neo, but not SH-SY5Y/Bcl-xL cells, mitochondrial fragmentation elicited by GSK2578215A precedes an autophagic response. Furthermore, the overexpression of Bcl-xL protein restores the autophagic flux pathway disrupted by this inhibitor. SH-SY5Y/Neo, but not SH-SY5Y/Bcl-xL cells, responded to LRRK2 inhibition by an increase in the levels of acetylated tubulin, indicating that this was abrogated by Bcl-xL overexpression. This hyperacetylation of tubulin took place earlier than any of the above-mentioned events suggesting that it is involved in the autophagic flux interruption. Pre-treatment with tempol prevented the GSK2578215A-induced mitochondrial fragmentation, autophagy and the rise in acetylated tubulin in SH-SY5Y/Neo cells. Thus, these data support the notion that ROS act as a second messenger connexion between LRRK2 inhibition and these deleterious responses, which are markedly alleviated by the Bcl-xL-mediated ROS generation blockade.
- Published
- 2015
24. A tango for two: Dopamine and α‐synuclein synergy may explain nigrostriatal degeneration in Parkinson's disease
- Author
-
Vivian Chen and Sara Saez-Atienzar
- Subjects
Male ,0301 basic medicine ,Parkinson's disease ,Dopamine ,Mice, Transgenic ,Article ,Levodopa ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Caenorhabditis elegans ,Cells, Cultured ,Nigrostriatal degeneration ,Chemistry ,Dopaminergic Neurons ,Neurodegenerative Diseases ,medicine.disease ,Corpus Striatum ,Mice, Inbred C57BL ,Substantia Nigra ,030104 developmental biology ,Neurology ,alpha-Synuclein ,Female ,α synuclein ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Parkinson's disease (PD) is defined by the loss of dopaminergic neurons in the substantia nigra and the formation of Lewy body inclusions containing aggregated α-synuclein. Efforts to explain dopamine neuron vulnerability are hindered by the lack of dopaminergic cell death in α-synuclein transgenic mice. To address this, we manipulated both dopamine levels and α-synuclein expression. Nigrally targeted expression of mutant tyrosine hydroxylase with enhanced catalytic activity increased dopamine levels without damaging neurons in non-transgenic mice. In contrast, raising dopamine levels in mice expressing human A53T mutant α-synuclein induced progressive nigrostriatal degeneration and reduced locomotion. Dopamine elevation in A53T mice increased levels of potentially toxic α-synuclein oligomers, resulting in conformationally and functionally modified species. Moreover, in genetically tractable Caenorhabditis elegans models, expression of α-synuclein mutated at the site of interaction with dopamine prevented dopamine-induced toxicity. These data suggest that a unique mechanism links two cardinal features of PD: dopaminergic cell death and α-synuclein aggregation.
- Published
- 2018
- Full Text
- View/download PDF
25. Autophagy as a Neuroprotective Mechanism Against 3-Nitropropionic Acid-Induced Cell Death
- Author
-
Sara Saez-Atienzar, Luis Bonet-Ponce, Maria F. Galindo, and Joaquín Jordán
- Subjects
Programmed cell death ,medicine.anatomical_structure ,Chemistry ,Neurodegeneration ,Cell ,Autophagy ,Mitophagy ,medicine ,Mitochondrion ,medicine.disease ,Neuroprotection ,Intracellular ,Cell biology - Abstract
The natural environmental toxin 3-Nitropropionic acid (3-NP) is used to induce situations somehow similar to those that take place in human patients suffering the progressive neurodegenerative illness known as Huntington’s disease (HD). Biochemical studies have shown reduced activities of complex II-III in the striatum of human HD brain tissues. 3-NP, structurally similar to succinate, is an inhibitor of succinate dehydrogenases or respiratory complex II. As a consequence, 3-NP stimulates Ca2 + release from mitochondria, decreases mitochondrial membrane potential, induces mitochondrial swelling, stimulates cytochrome c release from mitochondria and leads to a rapid decline of ATP levels. Autophagy, defined as the lysosomal digestion of a cell’s own cytoplasmic material, exerts its functions under conditions that require energy and use of intracellular nutrients. Under these conditions, autophagy shows a rapid increase, permitting the release of substrates used for the maintenance of ATP levels and the new synthesis of proteins that plays a fundamental role in stress adaptation. Autophagy is activated as an adaptive catabolic process in response to different forms of metabolic stress, including ATP depletion, calcium oxidative stress and mitochondrial disruption. Autophagy participates in the turnover of mitochondria, a strictly regulated process called mitophagy. So, autophagy maintaining cell metabolic balance plays an important role in cell fate. Indeed, autophagy is essential for the survival of neurons, since they cannot dilute the level of altered proteins and damaged organelles by means of cell division. We herein summarize and discuss the relevance of intracellular autophagic pathways activated by 3-NP, thus trying to shed light on the potential mechanism underlying the involvement of autophagy in HD model. Finally, we summarize the role of pharmacological modulators in changing 3-NP-induced autophagy and their effects on cell survival.
- Published
- 2015
- Full Text
- View/download PDF
26. Parkinson disease and clathrin coat dynamics at synapses, why not?
- Author
-
Andrew B. Singleton and Sara Saez-Atienzar
- Subjects
0301 basic medicine ,Phosphoric monoester hydrolases ,Nerve Tissue Proteins ,Parkinson Disease ,Biology ,Clathrin coat ,Clathrin ,Axons ,Phosphoric Monoester Hydrolases ,Article ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Neurology ,Mutation ,Synapses ,Mutation (genetic algorithm) ,biology.protein ,Humans ,Neurology (clinical) - Abstract
Synaptojanin 1 (SJ1) is a major presynaptic phosphatase that couples synaptic vesicle endocytosis to the dephosphorylation of PI(4,5)P2, a reaction needed for the shedding of endocytic factors from their membranes. While the role of SJ1’s 5-phosphatase module in this process is well established, the contribution of its Sac phosphatase domain, whose preferred substrate is PI4P, remains unclear. Recently a homozygous mutation in its Sac domain was identified in early-onset Parkinsonism patients. We show that mice carrying this mutation developed neurological manifestations similar to those of human patients. Synapses of these mice displayed endocytic defects and a striking accumulation of clathrin coated intermediates strongly implicating Sac domain’s activity in endocytic protein dynamics. Mutant brains had elevated auxilin (PARK19) and parkin (PARK2) levels. Moreover, dystrophic axonal terminal changes were selectively observed in dopaminergic axons in the dorsal striatum. These results strengthen evidence for a link between synaptic endocytic dysfunction and Parkinson’s disease.
- Published
- 2017
- Full Text
- View/download PDF
27. 6-Hydroxydopamine as Preclinical Model of Parkinson’s Disease
- Author
-
Maria E. Solesio, Sara Saez-Atienzar, Maria F. Galindo, and Joaquín Jordán
- Subjects
Hydroxydopamine ,Parkinson's disease ,business.industry ,medicine ,medicine.disease ,business ,Neuroscience - Published
- 2014
- Full Text
- View/download PDF
28. Pharmacology and Pathology of Superoxide Dismutases (SOD)
- Author
-
Sandra Atienzar-Aroca, Maria F. Galindo, Sara Saez-Atienzar, Joaquín Jordán, and Maria E. Solesio
- Subjects
Superoxide dismutase ,chemistry.chemical_compound ,chemistry ,biology ,Superoxide ,biology.protein ,Pharmacology ,Mitochondrion ,Neuroprotection - Published
- 2014
- Full Text
- View/download PDF
29. The LRRK2 inhibitor GSK2578215A induces protective autophagy in SH-SY5Y cells: involvement of Drp-1-mediated mitochondrial fission and mitochondrial-derived ROS signaling
- Author
-
Sara Saez-Atienzar, Luis Bonet-Ponce, Maria F. Galindo, José R. Blesa, Francisco J. Romero, Joaquín Jordán, and Michael P. Murphy
- Subjects
Autophagosome ,Dynamins ,Cancer Research ,Ubiquinone ,Immunology ,Aminopyridines ,Biology ,Mitochondrion ,Protein Serine-Threonine Kinases ,BAG3 ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Mitochondrial Dynamics ,GTP Phosphohydrolases ,Mitochondrial Proteins ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Organophosphorus Compounds ,Cell Line, Tumor ,Autophagy ,Humans ,RNA, Small Interfering ,MitoQ ,Aldehydes ,Benzodiazepinones ,Cell Biology ,LRRK2 ,Cell biology ,Mitochondria ,Oxidative Stress ,Pyrimidines ,chemistry ,Benzamides ,Mitochondrial fission ,RNA Interference ,Original Article ,Signal transduction ,Microtubule-Associated Proteins ,Signal Transduction - Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene have been associated with Parkinson's disease, and its inhibition opens potential new therapeutic options. Among the drug inhibitors of both wild-type and mutant LRRK2 forms is the 2-arylmethyloxy-5-subtitutent-N-arylbenzamide GSK257815A. Using the well-established dopaminergic cell culture model SH-SY5Y, we have investigated the effects of GSK2578215A on crucial neurodegenerative features such as mitochondrial dynamics and autophagy. GSK2578215A induces mitochondrial fragmentation of an early step preceding autophagy. This increase in autophagosome results from inhibition of fusion rather than increases in synthesis. The observed effects were shared with LRRK2-IN-1, a well-described, structurally distinct kinase inhibitor compound or when knocking down LRRK2 expression using siRNA. Studies using the drug mitochondrial division inhibitor 1 indicated that translocation of the dynamin-related protein-1 has a relevant role in this process. In addition, autophagic inhibitors revealed the participation of autophagy as a cytoprotective response by removing damaged mitochondria. GSK2578215A induced oxidative stress as evidenced by the accumulation of 4-hydroxy-2-nonenal in SH-SY5Y cells. The mitochondrial-targeted reactive oxygen species scavenger MitoQ positioned these species as second messengers between mitochondrial morphologic alterations and autophagy. Altogether, our results demonstrated the relevance of LRRK2 in mitochondrial-activated pathways mediating in autophagy and cell fate, crucial features in neurodegenerative diseases.
- Published
- 2013
30. Characterization of mitophagy in the 6-hydoxydopamine Parkinson's disease model
- Author
-
Maria E. Solesio, Maria F. Galindo, Joaquín Jordán, and Sara Saez-Atienzar
- Subjects
chemistry.chemical_classification ,Reactive oxygen species ,Autophagy ,Mitochondrial Degradation ,Mitophagy ,Parkinson Disease ,Mitochondrion ,Biology ,Toxicology ,Cell biology ,Cell Line ,chemistry.chemical_compound ,Disease Models, Animal ,chemistry ,Cell culture ,Animals ,Humans ,Mitochondrial fission ,Oxidopamine ,Reactive Oxygen Species - Abstract
In the present study, the activation of autophagy and its interaction with the mitochondrial fission machinery was investigated in an experimental model of Parkinson's disease. The addition of 50µM 6-hydroxydopamine (6-OHDA) to the dopaminergic cell line SH-SY5Y profoundly stimulated formation of autophagosomes within 12h. Under these conditions, mitochondrial fission was also activated in a sustained manner, but this occurred at earlier time points (after 3h). Upon 6-OHDA treatment, dynamin-related protein 1 (Drp1) transiently translocated to mitochondria, with increased levels of mitochondrial Drp1 being observed after 3 and 9h. Pharmacological inhibition of Drp1, through treatment with the mitochondrial-division inhibitor-1 (mdivi-1), resulted in the abrogation of mitochondrial fission and in a decrease of the number of autophagic cells. In addition, 6-OHDA failed to induce the expression of the proapoptotic protein Bax in total cellular extracts although it did induce its migration to mitochondria. In our model, Bax migrated later than Drp1. However, Drp1 inhibition did not block Bax migration. These results show that reactive oxygen species but not quinone derivates act as mediators of autophagy at an early stage of the process. 6-OHDA induces hydrogen peroxide production, which was placed upstream of mitochondrial fission, given that mdivi-1 did not abrogate this increase. Furthermore, the 6-OHDA-induced activation of autophagy was also suppressed by addition of the free radical scavengers TEMPOL and MnTBAP. This effect could be reproduced by the addition of hydrogen peroxide, but not with aged 6-OHDA. To our knowledge, this is the first detailed study highlighting the various mediators that are implicated in mitochondrial alterations and autophagy of cells in response to 6-OHDA.
- Published
- 2012
31. P.1.001 3-Nitropropionic acid induces autophagy by forming mitochondrial permeability transition pore, not by activating mitochondrial fission
- Author
-
J. Jordan, M.E. Solesio, M.F. Galindo, and Sara Saez-Atienzar
- Subjects
Pharmacology ,Virtual screening ,Chemistry ,Drug discovery ,Ligand (biochemistry) ,Fusion protein ,Psychiatry and Mental health ,Neurology ,Mitochondrial permeability transition pore ,Affinity chromatography ,Biophysics ,Pharmacology (medical) ,Mitochondrial fission ,Neurology (clinical) ,Biological Psychiatry ,G protein-coupled receptor - Abstract
and purified on an IMAC Nickel affinity column using a His-tag at the C terminus. Other expression systems that have been used for structural studies with GPCRs include yeast, E. coli and mammalian cells. Crystallisation screens are set up in a wide range of different detergent conditions. Two approaches are routinely used: vapour diffusion in short chain detergents and lipidic cubic phase (LCP) crystallisation. The lipidic cubic phase is a lipid matrix which produces a more native, membranelike environment. Extensive screening and optimisation of crystallisation conditions is then required to obtain the best diffracting crystals. Crystallisation in LCP is frequently facilitated by making fusion proteins with the GPCRwhich increase crystallisation contacts. For example T4 lysozyme may be fused into the third intracellular loop of the receptor. Antibodies may also be included as crystallisation chaperones. The resulting GPCR crystals are small and must be taken to specialised microfocussed X-ray beams at the synchrotron. Selection of the ligand for co-crystallisation studies is also important. In the absence of thermostabilising mutations a high affinity stabilising ligand is required to obtain crystals. The presence of thermostabilising mutations reduces this requirement and enables multiple co-crystal structures of weaker ligands to be obtained. This is important when using co-structures during the lead optimisation stage of drug discovery. Once the structure is solved a model of the receptor structure is obtained and this is used for virtual screening and structure based drug discovery. X-ray structures are highly enabling for GPCR drug discovery as they allow drug candidates to be designed which fit efficiently into the ligand binding pocket [3]. Compounds can be optimised for affinity, kinetics and selectivity using structure based approaches. Heptares have applied this approach to a range of CNS discovery projects including an adenosine A2A antagonist for the treatment of Parkinson’s disease, an orexin receptor antagonist for the treatment of sleep disorders and a highly selective muscarinic M1 agonist for the treatment of Alzheimer’s disease.
- Published
- 2013
- Full Text
- View/download PDF
32. Glutathione-sensitive nanoplatform for monitored intracellular delivery and controlled release of Camptothecin
- Author
-
Carlos Muniesa, Manuel Quesada, Víctor Vicente, Sara Saez-Atienzar, José R. Blesa, P. Botella, Ibane Abasolo, and Yolanda Fernández
- Subjects
Folate ,Cancer cells ,General Chemical Engineering ,Carbon nanotubes ,Tripeptide ,Gene delivery ,Endocytosis ,chemistry.chemical_compound ,In vivo ,medicine ,Mesopourus silica nanoparticles ,Disulfide bonds ,Chemistry ,Agents ,General Chemistry ,Glutathione ,Responsive nanoparticles ,Controlled release ,Biochemistry ,Drug delivery ,Biophysics ,Intracellular ,Camptothecin ,medicine.drug - Abstract
[EN] We report the design, synthesis, characterization and in vitro testing of a novel nanodrug based on a covalent linking model that allows intracellular controlled release of the pharmaceutical payload. A new synthetic strategy is implemented by direct coupling of as-synthesized (pyridin-2-yldisulfanyl)alkyl carbonate derivatives of camptothecin (CPT) with thiol groups of silica hybrid nanoparticles containing a non-porous core and a mesoporous shell. Upon reaction with thiols in physiological conditions, disulfide bridge cleavage occurs, releasing the naked drug after an intramolecular cyclization mechanism. Additional incorporation of a fluorophore into particles core facilitates imaging at the subcellular level for the monitoring of uptake and delivery. Confocal microscopy experiments in HeLa cervix cancer cells confirms that nanoparticles enter the cells by endocytosis but are able to escape from endo-lysosomes and enter the cytosolic compartment to release their cargo. The incorporation to cells of L-buthionine-sulfoximine, a glutathione inhibitor allows concluding that the intracellular releasing mechanism is mainly driven by the reducing activity of this tripeptide. This camptothecin nanoplatform shows the same cytotoxic activity than the free drug and is clearly superior to those release systems depending on enzymatic hydrolysis (as determined by calculation of the IC50 ratios)., This work was financially supported by "Comision Interministerial de Ciencia y Tecnologia" of Spain (projects CSD2009-00050 and MAT2012-39290-C02-02), and grants from CIBER-BBN (NanoMets Intramural Grant) "Fondo de Investigaciones Sanitarias - Instituto de Salud Carlos III" (PI080771) y "Universidad Catolica de Valencia San Vicente Martir" (PI2011-011-010). CM thanks the Spanish "Ministerio de Economia y Competitividad" for a FPU Ph.D. studentship (AP2008-02851). SSA thanks the "Universidad Catolica de Valencia San Vicente Martir" for a Ph.D. studentship.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.