Network


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

Hotspot


Dive into the research topics where Juan A. Godoy is active.

Publication


Featured researches published by Juan A. Godoy.


Molecular Psychiatry | 2003

Activation of Wnt signaling rescues neurodegeneration and behavioral impairments induced by β -amyloid fibrils

G. V. De Ferrari; Marcelo A. Chacón; María Inés Barría; Jose Luis Garrido; Juan A. Godoy; Gonzalo H. Olivares; Ariel E. Reyes; Anne Alvarez; Miguel Bronfman; Nibaldo C. Inestrosa

Alzheimers disease (AD) is a progressive neurodegenerative disorder, which is probably caused by the cytotoxic effect of the amyloid β-peptide (Aβ). We report here molecular changes induced by Aβ, both in neuronal cells in culture and in rats injected in the dorsal hippocampus with preformed Aβ fibrils, as an in vivo model of the disease. Results indicate that in both systems, Aβ neurotoxicity resulted in the destabilization of endogenous levels of β-catenin, a key transducer of the Wnt signaling pathway. Lithium chloride, which mimics Wnt signaling by inhibiting glycogen synthase kinase-3β promoted the survival of post-mitotic neurons against Aβ neurotoxicity and recovered cytosolic β-catenin to control levels. Moreover, the neurotoxic effect of Aβ fibrils was also modulated with protein kinase C agonists/inhibitors and reversed with conditioned medium containing the Wnt-3a ligand. We also examined the spatial memory performance of rats injected with preformed Aβ fibrils in the Morris water maze paradigm, and found that chronic lithium treatment protected neurodegeneration by rescuing β-catenin levels and improved the deficit in spatial learning induced by Aβ. Our results are consistent with the idea that Aβ-dependent neurotoxicity induces a loss of function of Wnt signaling components and indicate that lithium or compounds that mimic this signaling cascade may be putative candidates for therapeutic intervention in Alzheimers patients.


Journal of Biological Chemistry | 2008

Wnt-7a Modulates the Synaptic Vesicle Cycle and Synaptic Transmission in Hippocampal Neurons

Waldo Cerpa; Juan A. Godoy; Iván E. Alfaro; Ginny G. Farías; María J. Metcalfe; Rodrigo A. Fuentealba; Christian Bonansco; Nibaldo C. Inestrosa

Wnt signaling is essential for neuronal development and the maintenance of the developing nervous system. Recent studies indicated that Wnt signaling modulates long term potentiation in adult hippocampal slices. We report here that different Wnt ligands are present in hippocampal neurons of rat embryo and adult rat, including Wnt-4, -5a, -7a, and -11. Wnt-7a acts as a canonical Wnt ligand in rat hippocampal neurons, stimulates clustering of presynaptic proteins, and induces recycling and exocytosis of synaptic vesicles as studied by FM dyes. Wnt-3a has a moderate effect on recycling of synaptic vesicles, and no effect of Wnt-1 and Wnt-5a was detected. Electrophysiological analysis on adult rat hippocampal slices indicates that Wnt-7a, but not Wnt-5a, increases neurotransmitter release in CA3-CA1 synapses by decreasing paired pulse facilitation and increasing the miniature excitatory post-synaptic currents frequency. These results indicate that the presynaptic function of rat hippocampal neurons is modulated by the canonical Wnt signaling.


The FASEB Journal | 2002

Protein kinase C inhibits amyloid β peptide neurotoxicity by acting on members of the Wnt pathway

Jose Luis Garrido; Juan A. Godoy; Alejandra R. Alvarez; Miguel Bronfman; Nibaldo C. Inestrosa

Current evidence supports the notion that the amyloid β‐peptide (Aβ) plays a major role in the neurotoxicity observed in the brain in Alzheimers disease. However, the signal transduction mechanisms involved still remain unknown. In the present work, we analyzed the effect of protein kinase C (PKC) on some members of the Wnt signaling pathway and its implications for Aβ neurotoxicity. Activation of PKC by phorbol 12‐myristate 13‐acetate protected rat hippocampal neurons from Aβ toxicity. This effect was accomplished by inhibition of glycogen synthase kinase‐3β (GSK‐3β) activity, which led to the accumulation of cytoplasmic β‐catenin and transcriptional activation via β‐catenin/T‐cell factor/lymphoid enhancer factor‐1 (TCF/LEF1) of Wnt target genes, which in the present study were engrailed‐1 (en‐1) and cyclin D1 (cycD1). In contrast, inhibition of Ca2+‐dependent PKC isoforms activated GSK‐3β and offered no protection from Aβ neurotoxicity. Wnt‐3a and lithium salts, classical activators of the Wnt pathway, mimicked PKC activation. Our results suggest that regulation of members of the Wnt signaling pathway by Ca2+‐dependent PKC isoforms may be important in controlling the neurotoxic process induced by Aβ.


Journal of Biological Chemistry | 2009

Wnt-5a/JNK Signaling Promotes the Clustering of PSD-95 in Hippocampal Neurons

Ginny G. Farías; Iván E. Alfaro; Waldo Cerpa; Catalina Grabowski; Juan A. Godoy; Christian Bonansco; Nibaldo C. Inestrosa

During the formation of synapses, specific regions of pre- and postsynaptic cells associate to form a single functional transmission unit. In this process, synaptogenic factors are necessary to modulate pre- and postsynaptic differentiation. In mammals, different Wnt ligands operate through canonical and non-canonical Wnt pathways, and their precise functions to coordinate synapse structure and function in the mature central nervous system are still largely unknown. Here, we studied the effect of different Wnt ligands on postsynaptic organization. We found that Wnt-5a induces short term changes in the clustering of PSD-95, without affecting its total levels. Wnt-5a promotes the recruitment of PSD-95 from a diffuse dendritic cytoplasmic pool to form new PSD-95 clusters in dendritic spines. Moreover, Wnt-5a acting as a non-canonical ligand regulates PSD-95 distribution through a JNK-dependent signaling pathway, as demonstrated by using the TAT-TI-JIP peptide in mature hippocampal neurons. Finally, using adult rat hippocampal slices, we found that Wnt-5a modulates glutamatergic synaptic transmission through a postsynaptic mechanism. Our studies indicate that the Wnt-5a/JNK pathway modulates the postsynaptic region of mammalian synapse directing the clustering and distribution of the physiologically relevant scaffold protein, PSD-95.


Journal of Biological Chemistry | 2005

Peroxisomal Proliferation Protects from β-Amyloid Neurodegeneration

Manuel J. Santos; Rodrigo A. Quintanilla; Andrés Toro; Rodrigo Grandy; Margarita C. Dinamarca; Juan A. Godoy; Nibaldo C. Inestrosa

Alzheimer disease is a neurodegenerative process that leads to severe cognitive impairment as a consequence of selective death of neuronal populations. The molecular pathogenesis of Alzheimer disease involves the participation of the β-amyloid peptide (Aβ) and oxidative stress. We report here that peroxisomal proliferation attenuated Aβ-dependent toxicity in hippocampal neurons. Pretreatment with Wy-14.463 (Wy), a peroxisome proliferator, prevent the neuronal cell death and neuritic network loss induced by the Aβ peptide. Moreover, the hippocampal neurons treated with this compound, showed an increase in the number of peroxisomes, with a concomitant increase in catalase activity. Additionally, we evaluate the Wy protective effect on β-catenin levels, production of intracellular reactive oxygen species, cytoplasmic calcium uptake, and mitochondrial potential in hippocampal neurons exposed to H2 O2 and Aβ peptide. Results show that the peroxisomal proliferation prevents β-catenin degradation, reactive oxygen species production, cytoplasmic calcium increase, and changes in mitochondrial viability. Our data suggest, for the first time, a direct link between peroxisomal proliferation and neuroprotection from Aβ-induced degenerative changes.


The Journal of Neuroscience | 2010

Wnt-5a Modulates Recycling of Functional GABAA Receptors on Hippocampal Neurons

Loreto Cuitino; Juan A. Godoy; Ginny G. Farías; Andrés Couve; Christian Bonansco; Marco Fuenzalida; Nibaldo C. Inestrosa

GABAA receptors (GABAA-Rs) play a significant role in mediating fast synaptic inhibition and it is the main inhibitory receptor in the CNS. The role of Wnt signaling in coordinating synapse structure and function in the mature CNS is poorly understood. In previous studies we found that Wnt ligands can modulate excitatory synapses through remodeling both presynaptic and postsynaptic regions. In this current study we provide evidence for the effect of Wnt-5a on postsynaptic GABAA-Rs. We observed that Wnt-5a induces surface expression and maintenance of this receptor in the neuronal membrane. The evoked IPSC recordings in rat hippocampal slice indicate that Wnt-5a can regulates postsynaptically the hippocampal inhibitory synapses. We found also that Wnt-5a: (a) induces the insertion and clustering of GABAA-Rs in the membrane; (b) increases the amplitude of GABA-currents due exclusively to postsynaptic mechanisms; (c) does not affect the endocytic process, but increases the receptor recycling. Finally, all these effects on the GABAA-Rs are mediated by the activation of calcium/calmodulin-dependent kinase II (CaMKII). Therefore, we postulate that Wnt-5a, by activation of CaMKII, induces the recycling of functional GABAA-Rs on the mature hippocampal neurons.


The Journal of Neuroscience | 2007

Wnt-7a Induces Presynaptic Colocalization of α7-Nicotinic Acetylcholine Receptors and Adenomatous Polyposis Coli in Hippocampal Neurons

Ginny G. Farías; Ana Sofia Valles; Marcela Colombres; Juan A. Godoy; Enrique M. Toledo; Ronald J. Lukas; Francisco J. Barrantes; Nibaldo C. Inestrosa

Nicotinic acetylcholine receptors (nAChRs) contribute significantly to hippocampal function. α7-nAChRs are present in presynaptic sites in hippocampal neurons and may influence transmitter release, but the factors that determine their presynaptic localization are unknown. We report here that Wnt-7a, a ligand active in the canonical Wnt signaling pathway, induces dissociation of the adenomatous polyposis coli (APC) protein from the β-catenin cytoplasmic complex and the interaction of APC with α7-nAChRs in hippocampal neurons. Interestingly, Wnt-7a induces the relocalization of APC to membranes, clustering of APC in neurites, and coclustering of APC with different, presynaptic protein markers. Wnt-7a also increases the number and size of coclusters of α7-nAChRs and APC in presynaptic terminals. These short-term changes in α7-nAChRs occur in the few minutes after ligand exposure and involve translocation to the plasma membrane without affecting total receptor levels. Longer-term exposure to Wnt-7a increases nAChR α7 subunit levels in an APC-independent manner and increases clusters of α7-nAChRs in neurites via an APC-dependent process. Together, these results demonstrate that stimulation through the canonical Wnt pathway regulates the presynaptic localization of APC and α7-nAChRs with APC serving as an intermediary in the α7-nAChR relocalization process. Modulation by Wnt signaling may be essential for α7-nAChR expression and function in synapses.


Molecular Neurodegeneration | 2010

Wnt-5a occludes Aβ oligomer-induced depression of glutamatergic transmission in hippocampal neurons

Waldo Cerpa; Ginny G. Farías; Juan A. Godoy; Marco Fuenzalida; Christian Bonansco; Nibaldo C. Inestrosa

BackgroundSoluble amyloid-β (Aβ;) oligomers have been recognized to be early and key intermediates in Alzheimers disease (AD)-related synaptic dysfunction. Aβ oligomers block hippocampal long-term potentiation (LTP) and impair rodent spatial memory. Wnt signaling plays an important role in neural development, including synaptic differentiation.ResultsWe report here that the Wnt signaling activation prevents the synaptic damage triggered by Aβ oligomers. Electrophysiological analysis of Schaffer collaterals-CA1 glutamatergic synaptic transmission in hippocampal slices indicates that Wnt-5a increases the amplitude of field excitatory postsynaptic potentials (fEPSP) and both AMPA and NMDA components of the excitatory postsynaptic currents (EPSCs), without modifying the paired pulse facilitation (PPF). Conversely, in the presence of Aβ oligomers the fEPSP and EPSCs amplitude decreased without modification of the PPF, while the postsynaptic scaffold protein (PSD-95) decreased as well. Co-perfusion of hippocampal slices with Wnt-5a and Aβ oligomers occludes against the synaptic depression of EPSCs as well as the reduction of PSD-95 clusters induced by Aβ oligomers in neuronal cultures. Taken together these results indicate that Wnt-5a and Aβ oligomers inversely modulate postsynaptic components.ConclusionThese results indicate that post-synaptic damage induced by Aβ oligomers in hippocampal neurons is prevented by non-canonical Wnt pathway activation.


Acta Neurologica Scandinavica | 2000

Acetylcholinesterase-amyloid-β-peptide interaction and Wnt signaling involvement in Aβ neurotoxicity

Nibaldo C. Inestrosa; Anne Alvarez; Juan A. Godoy; Ariel E. Reyes; G. V. De Ferrari

Previous studies have indicated that acetylcholinesterase (AChE) promotes amyloid‐β‐peptide (Aβ) fibril formation and AChE‐Aβ complexes increase Aβ‐dependent neurotoxicity. Here we present evidence for the: i) identification of the AChE motif that promotes amyloid formation, ii) in vivo effect of AChE on brain plaque formation, and iii) connection between AChE‐Aβ neurotoxicity and the Wnt signal transduction pathway. Computer modeling, stereotaxic infusions and cell biological techniques were used to study the above problems. Results indicated that a 3.4 kDa AChE peptide promotes Aβ fibril formation. AChE infusion into rat hippocampus determines the appearance of anti‐Aβ and thioflavine‐S positive plaques, and AChE‐Aβ toxicity on hippocampal cultures was blocked by lithium, an activator of the Wnt cascade. We suggest that AChE‐Aβ/Aβ dependent neurotoxicity may result in loss of function of Wnt signaling components, and open the possibility that lithium may be considered as a candidate for therapeutic intervention in Alzheimers disease pathology.


PLOS ONE | 2013

Peroxisome Proliferator-Activated Receptor (PPAR) γ and PPARα Agonists Modulate Mitochondrial Fusion-Fission Dynamics: Relevance to Reactive Oxygen Species (ROS)-Related Neurodegenerative Disorders?

Juan M. Zolezzi; Carmen Silva-Alvarez; Daniela Ordenes; Juan A. Godoy; Francisco J. Carvajal; Manuel J. Santos; Nibaldo C. Inestrosa

Recent studies showed that the activation of the retinoid X receptor, which dimerizes with peroxisome proliferator-activated receptors (PPARs), leads to an enhanced clearance of Aβ from the brain of transgenic mice model of Alzheimer’s disease (AD), because an increased expression of apolipoprotein E and it main transporters. However, the effects observed must involve additional underlying mechanisms that have not been yet explored. Several studies conducted in our laboratory suggest that part of the effects observed for the PPARs agonist might involves mitochondrial function and, particularly, mitochondrial dynamics. In the present study we assessed the effects of oxidative stress challenge on mitochondrial morphology and mitochondrial dynamics-related proteins in hippocampal neurons. Using immunofluorescence, we evaluated the PPARγ co-activator 1α (PGC-1α), dynamin related protein 1 (DRP1), mitochondrial fission protein 1 (FIS1), and mitochondrial length, in order to determine if PPARs agonist pre-treatment is able to protect mitochondrial population from hippocampal neurons through modulation of the mitochondrial fusion-fission events. Our results suggest that both a PPARγ agonist (ciglitazone) and a PPARα agonist (WY 14.643) are able to protect neurons by modulating mitochondrial fusion and fission, leading to a better response of neurons to oxidative stress, suggesting that a PPAR based therapy could acts simultaneously in different cellular components. Additionally, our results suggest that PGC-1α and mitochondrial dynamics should be further studied in future therapy research oriented to ameliorate neurodegenerative disorders, such as AD.

Collaboration


Dive into the Juan A. Godoy's collaboration.

Top Co-Authors

Avatar

Nibaldo C. Inestrosa

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ginny G. Farías

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Waldo Cerpa

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar

Juvenal A. Ríos

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francisco J. Carvajal

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar

Macarena S. Arrázola

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nady Braidy

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge