Network


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

Hotspot


Dive into the research topics where Juan Carlos Arévalo is active.

Publication


Featured researches published by Juan Carlos Arévalo.


Cellular and Molecular Life Sciences | 2006

Neurotrophin signaling: many exciting surprises!

Juan Carlos Arévalo; Synphen H. Wu

Abstract.Neurotrophins are growth factors implicated in the development and maintenance of different neuronal populations in the nervous system. Neurotrophins bind to two sets of receptors, Trk receptor tyrosine kinases and the p75NTR receptor, to activate several different signaling pathways that mediate various biological functions. While Trk receptor activation has been well-studied and triggers the well-characterized Ras/Rap-MAPK, PI3K-Akt, and PLCγ-PKC cascades, p75NTR signaling is more complex, and its in vivo significance has not yet been completely determined. In the last few years, p75NTR has received much attention mainly due to recent findings describing pro-neurotrophins as new ligands for the receptor and the ability of the receptor to form different complexes with other transmembrane proteins. This review will update the neurotrophin signaling pathways known for Trk receptors to include newly identified Trk-interacting molecules and will address surprising new findings that suggest a role for p75NTR in different receptor complexes and functions.


Neuron | 2006

Cell Survival through Trk Neurotrophin Receptors Is Differentially Regulated by Ubiquitination

Juan Carlos Arévalo; Janelle Waite; Rithwick Rajagopal; Mercedes Beyna; Zhe-Yu Chen; Francis S. Lee; Moses V. Chao

Specificity of neurotrophin factor signaling is dictated through the action of Trk receptor tyrosine kinases. Once activated, Trk receptors are internalized and targeted for degradation. However, the mechanisms implicated in this process are incompletely understood. Here we report that the Trk receptors are multimonoubiquitinated in response to neurotrophins. We have identified an E3 ubiquitin ligase, Nedd4-2, that associates with the TrkA receptor and is phosphorylated upon NGF binding. The binding of Nedd4-2 to TrkA through a PPXY motif leads to the ubiquitination and downregulation of TrkA. Activated TrkA receptor levels and the survival of NGF-dependent sensory neurons, but not BDNF-dependent sensory neurons, are directly influenced by Nedd4-2 expression. Unexpectedly, Nedd4-2 does not bind or ubiquitinate related TrkB receptors, due to the lack of a consensus PPXY motif. Our results indicate that Trk neurotrophin receptors are differentially regulated by ubiquitination to modulate the survival of neurons.


American Journal of Pathology | 2005

Pro-NGF isolated from the human brain affected by Alzheimer's disease induces neuronal apoptosis mediated by p75NTR.

Carlos Pedraza; Petar Podlesniy; Noemí Vidal; Juan Carlos Arévalo; Ramee Lee; Barbara L. Hempstead; Isidre Ferrer; Montse Iglesias; Carme Espinet

The pro-form of nerve growth factor (pro-NGF) has been shown to be a high affinity ligand for p75NTR and to induce apoptosis through this receptor. It has been reported that pro-NGF, rather than mature NGF, is the predominant form of this neurotrophin in human brain. In the present work we studied the potential involvement of pro-NGF purified from human brains affected by Alzheimers disease (AD), where it is especially abundant, in the neuronal apoptosis observed in this disease. Western blot analysis of human brain tissue showed the existence of several pro-NGF forms. Some of these pro-NGF forms were significantly increased in AD brain cortex in a disease stage-dependent manner. Pro-NGF, purified by chromatography from human AD brains, induced apoptotic cell death in sympathetic neurons and in a p75NTR stably transfected cell line. Blocking p75NTR in cell culture abolished neuronal apoptosis caused by pro-NGF. p75NTR-transfected cells underwent apoptosis in the presence of pro-NGF while control wild-type cells did not. Taken together, these results indicate that pro-NGF purified from AD human brains can induce apoptosis in neuronal cell cultures through its interaction with the p75NTR receptor.


The EMBO Journal | 2004

A unique pathway for sustained neurotrophin signaling through an ankyrin‐rich membrane‐spanning protein

Juan Carlos Arévalo; Hiroko Yano; Kenneth K. Teng; Moses V. Chao

A major question in cell biology is how molecular specificity is achieved by different growth factor receptors that activate apparently identical signaling events. For the neurotrophin family, a distinguishing feature is the ability to maintain a prolonged duration of signal transduction. However, the mechanisms by which neurotrophin receptors assemble such a sustained signaling complex are not understood. Here we report that an unusual ankyrin‐rich transmembrane protein (ARMS+kidins220) is closely associated with Trk receptor tyrosine kinases, and not the EGF receptor. This association requires interactions between transmembrane domains of Trk and ARMS. ARMS is rapidly tyrosine phosphorylated after binding of neurotrophins to Trk receptors and provides a docking site for the CrkL–C3G complex, resulting in Rap1‐dependent sustained ERK activation. Accordingly, disruption of Trk–ARMS or the ARMS–CrkL interaction with dominant‐negative ARMS mutants, or treatment with small interference RNA against ARMS substantially reduce neurotrophin‐elicited signaling to ERK, but without any effect upon Ras or Akt activation. These findings suggest that ARMS acts as a major and neuronal‐specific platform for prolonged MAP kinase signaling by neurotrophins.


Molecular and Cellular Biology | 2000

TrkA Immunoglobulin-Like Ligand Binding Domains Inhibit Spontaneous Activation of the Receptor

Juan Carlos Arévalo; Blanca Conde; Barbara L. Hempstead; Moses V. Chao; Dionisio Martin-Zanca; Pilar Pérez

ABSTRACT The extracellular region of the nerve growth factor (NGF) receptor, TrkA, contains two immunoglobulin (Ig)-like domains that are required for specific ligand binding. We have investigated the possible role of these two Ig-like domains in receptor dimerization and activation by using different mutants of the TrkA extracellular region. Deletions of each Ig-like domain, of both, and of the entire extracellular region were made. To probe the structural constraints on ligand-independent receptor dimerization, chimeric receptors were generated by swapping the Ig-like domains of the TrkA receptor for the third or fourth Ig-like domain of c-Kit. We also introduced single-amino-acid changes in conserved residues within the Ig-like domains of TrkA. Most of these TrkA variants did not bind NGF, and their expression in PC12nnr5 cells, which lack endogenous TrkA, promoted ligand-independent neurite outgrowth. Some TrkA mutant receptors induced malignant transformation of Rat-1 cells, as assessed by measuring proliferation in the absence of serum, anchorage-independent growth, and tumorigenesis in nude mice. These mutants exhibited constitutive phosphorylation and spontaneous dimerization consistent with their biological activities. Our data suggest that spontaneous dimerization of TrkA occurs when the structure of the Ig-like domains is altered, implying that the intact domains inhibit receptor dimerization in the absence of NGF.


Journal of Biological Chemistry | 2006

Identification of a Switch in Neurotrophin Signaling by Selective Tyrosine Phosphorylation

Juan Carlos Arévalo; Daniela B. Pereira; Hiroko Yano; Kenneth K. Teng; Moses V. Chao

Neurotrophins, such as nerve growth factor and brain-derived neurotrophic factor, activate Trk receptor tyrosine kinases through receptor dimerization at the cell surface followed by autophosphorylation and recruitment of intracellular signaling molecules. The intracellular pathways used by neurotrophins share many common protein substrates that are used by other receptor tyrosine kinases (RTK), such as Shc, Grb2, FRS2, and phospholipase C-γ. Here we describe a novel RTK mechanism that involves a 220-kilodalton membrane tetraspanning protein, ARMS/Kidins220, which is rapidly tyrosine phosphorylated in primary neurons after neurotrophin treatment. ARMS/Kidins220 undergoes multiple tyrosine phosphorylation events and also serine phosphorylation by protein kinase D. We have identified a single tyrosine (Tyr1096) phosphorylation event in ARMS/Kidins220 that plays a critical role in neurotrophin signaling. A reassembled complex of ARMS/Kidins220 and CrkL, an upstream component of the C3G-Rap1-MAP kinase cascade, is SH3-dependent. However, Tyr1096 phosphorylation enables ARMS/Kidins220 to recruit CrkL through its SH2 domain, thereby freeing the CrkL SH3 domain to engage C3G for MAP kinase activation in a neurotrophin dependent manner. Accordingly, mutation of Tyr1096 abolished CrkL interaction and sustained MAPK kinase activity, a response that is not normally observed in other RTKs. Therefore, Trk receptor signaling involves an inducible switch mechanism through an unconventional substrate that distinguishes neurotrophin action from other growth factor receptors.


Journal of Neuroscience Research | 2004

Ternary Complex with Trk, p75, and an Ankyrin-Rich Membrane Spanning Protein

Mi-Sook Chang; Juan Carlos Arévalo; Moses V. Chao

Neurotrophins play many critical roles in regulating neuronal plasticity, survival, and differentiation in the nervous system. Neurotrophins recognize two different receptors, the Trk receptor tyrosine kinase and the p75 neurotrophin receptor, which are associated closely. Several adaptor proteins are associated with each receptor. An ankyrin‐rich membrane spanning protein (ARMS), originally identified as a substrate for protein kinase D (Kidins220) and as a p75 interacting protein, serves as a novel downstream target of Trk receptor tyrosine kinases. Kidins220/ARMS is co‐expressed frequently with Trk and p75 and represents the only membrane‐associated protein known to interact with both receptors. We report here that a ternary complex can be formed between Trk, p75, and Kidins220/ARMS. The extracellular domains of the TrkA and the p75 receptors are necessary for their association, whereas the juxtamembrane region of p75 was responsible for the interaction with Kidins220/ARMS. Interestingly, increasing the level of Kidins220/ARMS expression resulted in a decreased association of TrkA with p75. These findings thus suggest that Kidins220/ARMS plays an important role in regulating interactions between Trk and p75 neurotrophin receptors.


Journal of Cell Biology | 2004

The p75NTR-interacting protein SC1 inhibits cell cycle progression by transcriptional repression of cyclin E.

Alexandra Chittka; Juan Carlos Arévalo; Maria Rodriguez-Guzman; Pilar Pérez; Moses V. Chao; Michael Sendtner

Schwann cell factor 1 (SC1), a p75 neurotrophin receptor–interacting protein, is a member of the positive regulatory/suppressor of variegation, enhancer of zeste, trithorax (PR/SET) domain-containing zinc finger protein family, and it has been shown to be regulated by serum and neurotrophins. SC1 shows a differential cytoplasmic and nuclear distribution, and its presence in the nucleus correlates strongly with the absence of bromodeoxyuridine (BrdU) in these nuclei. Here, we investigated potential transcriptional activities of SC1 and analyzed the function of its various domains. We show that SC1 acts as a transcriptional repressor when it is tethered to Gal4 DNA-binding domain. The repressive activity requires a trichostatin A–sensitive histone deacetylase (HDAC) activity, and SC1 is found in a complex with HDACs 1, 2, and 3. Transcriptional repression exerted by SC1 requires the presence of its zinc finger domains and the PR domain. Additionally, these two domains are involved in the efficient block of BrdU incorporation by SC1. The zinc finger domains are also necessary to direct SC1s nuclear localization. Lastly, SC1 represses the promoter of a promitotic gene, cyclin E, suggesting a mechanism for how growth arrest is regulated by SC1.


Developmental Neurobiology | 2009

Ankyrin Repeat-rich Membrane Spanning/Kidins220 protein regulates dendritic branching and spine stability in vivo.

Synphen H. Wu; Juan Carlos Arévalo; Federica Sarti; Lino Tessarollo; Wen-Biao Gan; Moses V. Chao

The development of nervous system connectivity depends upon the arborization of dendritic fields and the stabilization of dendritic spine synapses. It is well established that neuronal activity and the neurotrophin BDNF modulate these correlated processes. However, the downstream mechanisms by which these extrinsic signals regulate dendritic development and spine stabilization are less well known. Here we report that a substrate of BDNF signaling, the Ankyrin Repeat‐rich Membrane Spanning (ARMS) protein or Kidins220, plays a critical role in the branching of cortical and hippocampal dendrites and in the turnover of cortical spines. In the barrel somatosensory cortex and the dentate gyrus, regions where ARMS/Kidins220 is highly expressed, no difference in the complexity of dendritic arbors was observed in 1‐month‐old adolescent ARMS/Kidins220+/− mice compared to wild‐type littermates. However, at 3 months of age, young adult ARMS/Kidins220+/− mice exhibited decreased dendritic complexity. This suggests that ARMS/Kidins220 does not play a significant role in the initial formation of dendrites but, rather, is involved in the refinement or stabilization of the arbors later in development. In addition, at 1 month of age, the rate of spine elimination was higher in ARMS/Kidins220+/− mice than in wild‐type mice, suggesting that ARMS/Kidins220+/− levels regulate spine stability. Taken together, these data suggest that ARMS/Kidins220 is important for the growth of dendritic arbors and spine stability during an activity‐ and BDNF‐dependent period of development.


Journal of Cell Biology | 2005

α-Syntrophin regulates ARMS localization at the neuromuscular junction and enhances EphA4 signaling in an ARMS-dependent manner

Shuo Luo; Yu Chen; Kwok On Lai; Juan Carlos Arévalo; Stanley C. Froehner; Marvin E. Adams; Moses V. Chao; Nancy Y. Ip

EphA4 signaling has recently been implicated in the regulation of synapse formation and plasticity. In this study, we show that ankyrin repeat-rich membrane spanning (ARMS; also known as a kinase D–interacting substrate of 220 kD), a substrate for ephrin and neurotrophin receptors, was expressed in developing muscle and was concentrated at the neuromuscular junction (NMJ). Using yeast two-hybrid screening, we identified a PDZ (PSD-95, Dlg, ZO-1) domain protein, α-syntrophin, as an ARMS-interacting protein in muscle. Overexpression of α-syntrophin induced ARMS clustering in a PDZ domain–dependent manner. Coexpression of ARMS enhanced EphA4 signaling, which was further augmented by the presence of α-syntrophin. Moreover, the ephrin-A1–induced tyrosine phosphorylation of EphA4 was reduced in C2C12 myotubes after the blockade of ARMS and α-syntrophin expression by RNA interference. Finally, α-syntrophin–null mice exhibited a disrupted localization of ARMS and EphA4 at the NMJ and a reduced expression of ARMS in muscle. Altogether, our findings suggest that ARMS may play an important role in regulating postsynaptic signal transduction through the syntrophin-mediated localization of receptor tyrosine kinases such as EphA4.

Collaboration


Dive into the Juan Carlos Arévalo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Sanz-Merodio

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

E. Garcia

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Manuel Cestari

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Saray López-Benito

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Laura Calvo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Elena Garcia

Technical University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Begoña Anta

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Pilar Pérez

University of Salamanca

View shared research outputs
Researchain Logo
Decentralizing Knowledge