Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where René L. Vidal is active.

Publication


Featured researches published by René L. Vidal.


Human Molecular Genetics | 2012

Targeting the UPR transcription factor XBP1 protects against Huntington's disease through the regulation of FoxO1 and autophagy

René L. Vidal; Alicia Figueroa; Felipe A. Court; Peter Thielen; Claudia Molina; Craig Wirth; Benjamin Caballero; Roberta Kiffin; Juan Segura-Aguilar; Ana Maria Cuervo; Laurie H. Glimcher; Claudio Hetz

Mutations leading to expansion of a poly-glutamine track in Huntingtin (Htt) cause Huntingtons disease (HD). Signs of endoplasmic reticulum (ER) stress have been recently reported in animal models of HD, associated with the activation of the unfolded protein response (UPR). Here we have investigated the functional contribution of ER stress to HD by targeting the expression of two main UPR transcription factors, XBP1 and ATF4 (activating transcription factor 4), in full-length mutant Huntingtin (mHtt) transgenic mice. XBP1-deficient mice were more resistant to developing disease features, associated with improved neuronal survival and motor performance, and a drastic decrease in mHtt levels. The protective effects of XBP1 deficiency were associated with enhanced macroautophagy in both cellular and animal models of HD. In contrast, ATF4 deficiency did not alter mHtt levels. Although, XBP1 mRNA splicing was observed in the striatum of HD transgenic brains, no changes in the levels of classical ER stress markers were detected in symptomatic animals. At the mechanistic level, we observed that XBP1 deficiency led to augmented expression of Forkhead box O1 (FoxO1), a key transcription factor regulating autophagy in neurons. In agreement with this finding, ectopic expression of FoxO1 enhanced autophagy and mHtt clearance in vitro. Our results provide strong evidence supporting an involvement of XBP1 in HD pathogenesis probably due to an ER stress-independent mechanism involving the control of FoxO1 and autophagy levels.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Control of dopaminergic neuron survival by the unfolded protein response transcription factor XBP1

Pamela Valdés; Gabriela Mercado; René L. Vidal; Claudia Molina; Geoffrey Parsons; Felipe A. Court; Alexis Martínez; Danny Galleguillos; Donna Armentano; Bernard L. Schneider; Claudio Hetz

Significance The selective loss of dopaminergic neurons is characteristic of Parkinson disease (PD). Protein folding stress is a salient feature of PD. This study uncovers a previously undefined function of a major unfolded protein response (UPR) transcription factor (XBP1) in supporting the survival of nigral dopaminergic neurons at basal levels and under pathological conditions. Our results reveal an important role for a canonical UPR pathway in the maintenance of dopaminergic neuron proteostasis, which also could be relevant to understand the selective neuronal vulnerability observed in Parkinson disease. Parkinson disease (PD) is characterized by the selective loss of dopaminergic neurons of the substantia nigra pars compacta (SNpc). Although growing evidence indicates that endoplasmic reticulum (ER) stress is a hallmark of PD, its exact contribution to the disease process is not well understood. Here we report that developmental ablation of X-Box binding protein 1 (XBP1) in the nervous system, a key regulator of the unfolded protein response (UPR), protects dopaminergic neurons against a PD-inducing neurotoxin. This survival effect was associated with a preconditioning condition that resulted from induction of an adaptive ER stress response in dopaminergic neurons of the SNpc, but not in other brain regions. In contrast, silencing XBP1 in adult animals triggered chronic ER stress and dopaminergic neuron degeneration. Supporting this finding, gene therapy to deliver an active form of XBP1 provided neuroprotection and reduced striatal denervation in animals injected with 6-hydroxydopamine. Our results reveal a physiological role of the UPR in the maintenance of protein homeostasis in dopaminergic neurons that may help explain the differential neuronal vulnerability observed in PD.


Trends in Pharmacological Sciences | 2014

Targeting autophagy in neurodegenerative diseases.

René L. Vidal; Soledad Matus; Leslie Bargsted; Claudio Hetz

The most prevalent neurodegenerative disorders involve protein misfolding and the aggregation of specific proteins. Autophagy is becoming an attractive target to treat neurodegenerative disorders through the selective degradation of abnormally folded proteins by the lysosomal pathway. However, accumulating evidence indicates that autophagy impairment at different regulatory steps may contribute to the neurodegenerative process. Thus, a complex scenario is emerging where autophagy may play a dual role in neurodegenerative diseases by causing the downstream effect of promoting the degradation of misfolded proteins and an upstream effect where its deregulation perturbs global proteostasis, contributing to disease progression. Challenges in the future development of therapeutic strategies to target the autophagy pathway are discussed.


Cell Reports | 2016

Regulation of Memory Formation by the Transcription Factor XBP1

Gabriela Martínez; René L. Vidal; Pablo Mardones; Felipe G. Serrano; Alvaro O. Ardiles; Craig Wirth; Pamela Valdés; Peter Thielen; Bernard L. Schneider; Bredford Kerr; José L. Valdés; Adrian G. Palacios; Nibaldo C. Inestrosa; Laurie H. Glimcher; Claudio Hetz

Contextual memory formation relies on the induction of new genes in the hippocampus. A polymorphism in the promoter of the transcription factor XBP1 was identified as a risk factor for Alzheimers disease and bipolar disorders. XBP1 is a major regulator of the unfolded protein response (UPR), mediating adaptation to endoplasmic reticulum (ER) stress. Using a phenotypic screen, we uncovered an unexpected function of XBP1 in cognition and behavior. Mice lacking XBP1 in the nervous system showed specific impairment of contextual memory formation and long-term potentiation (LTP), whereas neuronal XBP1s overexpression improved performance in memory tasks. Gene expression analysis revealed that XBP1 regulates a group of memory-related genes, highlighting brain-derived neurotrophic factor (BDNF), a key component in memory consolidation. Overexpression of BDNF in the hippocampus reversed the XBP1-deficient phenotype. Our study revealed an unanticipated function of XBP1 in cognitive processes that is apparently unrelated to its role in ER stress.


Nature Communications | 2013

A failure in energy metabolism and antioxidant uptake precede symptoms of Huntington’s disease in mice

Acuña Ai; Esparza M; Kramm C; Beltrán Fa; Parra Av; Cepeda C; Toro Ca; René L. Vidal; Claudio Hetz; Concha; Brauchi S; Levine Ms; Castro Ma

Huntington’s disease has been associated with a failure in energy metabolism and oxidative damage. Ascorbic acid is a powerful antioxidant highly concentrated in the brain where it acts as a messenger, modulating neuronal metabolism. Using an electrophysiological approach in R6/2 HD slices, we observe an abnormal ascorbic acid flux from astrocytes to neurons, which is responsible for alterations in neuronal metabolic substrate preferences. Here using striatal neurons derived from knock-in mice expressing mutant huntingtin (STHdhQ cells), we study ascorbic acid transport. When extracellular ascorbic acid concentration increases, as occurs during synaptic activity, ascorbic acid transporter 2 (SVCT2) translocates to the plasma membrane, ensuring optimal ascorbic acid uptake for neurons. In contrast, SVCT2 from cells that mimic HD symptoms (dubbed HD cells) fails to reach the plasma membrane under the same conditions. We reason that an early impairment of ascorbic acid uptake in HD neurons could lead to early metabolic failure promoting neuronal death.


Biochemical and Biophysical Research Communications | 2012

AAV-mediated delivery of the transcription factor XBP1s into the striatum reduces mutant Huntingtin aggregation in a mouse model of Huntington’s disease

Amparo Zuleta; René L. Vidal; Donna Armentano; Geoffrey Parsons; Claudio Hetz

Huntingtons disease (HD) is caused by mutations that expand a polyglutamine region in the amino-terminal domain of Huntingtin (Htt), leading to the accumulation of intracellular inclusions and progressive neurodegeneration. Recent reports indicate the engagement of endoplasmic reticulum (ER) stress responses in human HD post mortem samples and animal models of the disease. Adaptation to ER stress is mediated by the activation of the unfolded protein response (UPR), an integrated signal transduction pathway that attenuates protein folding stress by controlling the expression of distinct transcription factors including X-Box binding protein 1 (XBP1). Here we targeted the expression of XBP1 on a novel viral-based model of HD. We delivered an active form of XBP1 locally into the striatum of adult mice using adeno-associated vectors (AAVs) and co-expressed this factor with a large fragment of mutant Htt as a fusion protein with RFP (Htt588(Q95)-mRFP) to directly visualize the accumulation of Htt inclusions in the brain. Using this approach, we observed a significant reduction in the accumulation of Htt588(Q95)-mRFP intracellular inclusion when XBP1 was co-expressed in the striatum. These results contrast with recent findings indicating a protective effect of XBP1 deficiency in neurodegeneration using knockout mice, and suggest a potential use of gene therapy strategies to manipulate the UPR in the context of HD.


Journal of Biological Chemistry | 2009

Dendritic Assembly of Heteromeric γ-Aminobutyric Acid Type B Receptor Subunits in Hippocampal Neurons

Omar A. Ramírez; René L. Vidal; Judith A. Tello; Karina J. Vargas; Stefan Kindler; Steffen Härtel; Andrés Couve

Understanding the mechanisms that control synaptic efficacy through the availability of neurotransmitter receptors depends on uncovering their specific intracellular trafficking routes. γ-Aminobutyric acid type B (GABAB) receptors (GABABRs) are obligatory heteromers present at dendritic excitatory and inhibitory postsynaptic sites. It is unknown whether synthesis and assembly of GABABRs occur in the somatic endoplasmic reticulum (ER) followed by vesicular transport to dendrites or whether somatic synthesis is followed by independent transport of the subunits for assembly and ER export throughout the somatodendritic compartment. To discriminate between these possibilities we studied the association of GABABR subunits in dendrites of hippocampal neurons combining live fluorescence microscopy, biochemistry, quantitative colocalization, and bimolecular fluorescent complementation. We demonstrate that GABABR subunits are segregated and differentially mobile in dendritic intracellular compartments and that a high proportion of non-associated intracellular subunits exist in the brain. Assembled heteromers are preferentially located at the plasma membrane, but blockade of ER exit results in their intracellular accumulation in the cell body and dendrites. We propose that GABABR subunits assemble in the ER and are exported from the ER throughout the neuron prior to insertion at the plasma membrane. Our results are consistent with a bulk flow of segregated subunits through the ER and rule out a post-Golgi vesicular transport of preassembled GABABRs.


The EMBO Journal | 2016

ALS-linked protein disulfide isomerase variants cause motor dysfunction

Ute Woehlbier; Alicia Colombo; Mirva J. Saaranen; Viviana Pérez; Jorge Ojeda; Fernando J. Bustos; Catherine Andreu; Mauricio Torres; Vicente Valenzuela; Danilo B. Medinas; Pablo Rozas; René L. Vidal; Rodrigo Lopez-Gonzalez; Johnny Salameh; Sara Fernández-Collemann; Natalia Muñoz; Soledad Matus; Ricardo Armisen; Alfredo I. Sagredo; Karina Palma; Thergiory Irrazabal; Sandra Almeida; Paloma Gonzalez-Perez; Mario Campero; Fen-Biao Gao; Pablo Henny; Brigitte van Zundert; Lloyd W. Ruddock; Miguel L. Concha; Juan Pablo Henríquez

Disturbance of endoplasmic reticulum (ER) proteostasis is a common feature of amyotrophic lateral sclerosis (ALS). Protein disulfide isomerases (PDIs) are ER foldases identified as possible ALS biomarkers, as well as neuroprotective factors. However, no functional studies have addressed their impact on the disease process. Here, we functionally characterized four ALS‐linked mutations recently identified in two major PDI genes, PDIA1 and PDIA3/ERp57. Phenotypic screening in zebrafish revealed that the expression of these PDI variants induce motor defects associated with a disruption of motoneuron connectivity. Similarly, the expression of mutant PDIs impaired dendritic outgrowth in motoneuron cell culture models. Cellular and biochemical studies identified distinct molecular defects underlying the pathogenicity of these PDI mutants. Finally, targeting ERp57 in the nervous system led to severe motor dysfunction in mice associated with a loss of neuromuscular synapses. This study identifies ER proteostasis imbalance as a risk factor for ALS, driving initial stages of the disease.


Autophagy | 2012

Crosstalk between the UPR and autophagy pathway contributes to handling cellular stress in neurodegenerative disease

René L. Vidal; Claudio Hetz

Huntington disease (HD) is caused by an extended polyglutamine [poly(Q)] stretch in the Huntingtin (HTT) protein, and is associated with the accumulation of intracellular protein aggregates, onset of progressive chorea, psychiatric symptoms and dementia. Although the mechanism underlying the pathological effects of mutant HTT (mHTT) remains highly controversial, accumulating evidence suggest that protein-folding stress at the endoplasmic reticulum (ER) may contribute to mHTT-mediated degeneration. ER stress is alleviated by the activation of an adaptive reaction known as the unfolded protein response (UPR), whereas chronic ER stress triggers apoptosis by the same pathway. However, most of the studies linking ER stress with HD in vivo are correlative. UPR signaling is initiated by the activation of at least three distinct stress sensors located at the ER membrane known as ERN1/IRE1α, EIF2AK3/PERK and ATF6. These stress sensors control the expression of specialized transcription factors that modulate the upregulation of a variety of target genes involved in folding, protein quality control, autophagy and protein synthesis.


Iubmb Life | 2013

Role of the unfolded protein response in organ physiology: Lessons from mouse models

Víctor Hugo Cornejo; Philippe Pihán; René L. Vidal; Claudio Hetz

The endoplasmic reticulum (ER) is a key subcellular compartment involved in the folding and maturation of around one‐third of the total proteome. Accumulation of misfolded proteins in the ER lumen engages a signal transduction pathway known as unfolded protein response (UPR) that feedback to recover ER homeostasis or to trigger apoptosis of irreversible damaged cells. The UPR is initiated by three main stress sensors including protein kinase RNA‐like ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol‐requiring protein 1α (IRE1α), which reprogram the genome through the control of downstream transcription factors. In this article, the authors have reviewed most relevant studies uncovering the physiological function of the UPR in different organs and tissues based on the phenotypes observed after genetic manipulation of the pathway in vivo. Biomedical applications of targeting the UPR on a disease context are also discussed.

Collaboration


Dive into the René L. Vidal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bredford Kerr

Centro de Estudios Científicos

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge