Network


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

Hotspot


Dive into the research topics where Eduardo Chavez is active.

Publication


Featured researches published by Eduardo Chavez.


Neuroscience | 2007

Role of inhibitor of apoptosis protein in gentamicin-induced cochlear hair cell damage

K. Tabuchi; Kwang Pak; Eduardo Chavez; Allen F. Ryan

Apoptotic cell death is considered to play a key role in gentamicin-induced cochlear hair cell loss. Inhibitor of apoptosis proteins (IAPs) are important regulators of apoptosis that can prevent activation of effector caspases. This study was designed to investigate the possible involvement of X-linked inhibitor of apoptosis protein (XIAP) in hair cell death due to gentamicin. Basal turn organ of Corti explants from postnatal day (p) p3 or p4 rats were maintained in tissue culture and were exposed to 35 muM gentamicin for up to 48 h. Effects of specific XIAP inhibitors on gentamicin-induced hair cell loss and caspase-3 activation were examined. XIAP inhibitors increased gentamicin-induced hair cell loss but an inactive analog had no effect. Caspase-3 activation was primarily observed at 36 or 48 h in gentamicin-treated hair cells, whereas caspase-3 activation peaked at 24-36 h when explants were treated with gentamicin and an XIAP inhibitor. The inhibitors alone had no effect on hair cells. Finally, a caspase-3 inhibitor decreased caspase-3 activation and hair cell loss induced by gentamicin and an XIAP inhibitor, but caspase-8 and -9 inhibitors did not. The results indicate that XIAP normally acts to decrease caspase-3 activation and hair cell loss during gentamicin ototoxicity, as part of a protective response to potentially damaging stimuli.


Molecular and Cellular Neuroscience | 2013

Glial cell line-derived neurotrophic factor (GDNF) induces neuritogenesis in the cochlear spiral ganglion via neural cell adhesion molecule (NCAM).

Sara Euteneuer; Kuo H. Yang; Eduardo Chavez; Anke Leichtle; Gabriele Loers; Adel Olshansky; Kwang Pak; Melitta Schachner; Allen F. Ryan

Glial cell line-derived neurotrophic factor (GDNF) increases survival and neurite extension of spiral ganglion neurons (SGNs), the primary neurons of the auditory system, via yet unknown signaling mechanisms. In other cell types, signaling is achieved by the GPI-linked GDNF family receptor α1 (GFRα1) via recruitment of transmembrane receptors: Ret (re-arranged during transformation) and/or NCAM (neural cell adhesion molecule). Here we show that GDNF enhances neuritogenesis in organotypic cultures of spiral ganglia from 5-day-old rats and mice. Addition of GFRα1-Fc increases this effect. GDNF/GFRα1-Fc stimulation activates intracellular PI3K/Akt and MEK/Erk signaling cascades as detected by Western blot analysis of cultures prepared from rats at postnatal days 5 (P5, before the onset of hearing) and 20 (P20, after the onset of hearing). Both cascades mediate GDNF stimulation of neuritogenesis, since application of the Akt inhibitor Wortmannin or the Erk inhibitor U0126 abolished GDNF/GFRα1-Fc stimulated neuritogenesis in P5 rats. Since cultures of P5 NCAM-deficient mice failed to respond by neuritogenesis to GDNF/GFRα1-Fc, we conclude that NCAM serves as a receptor for GDNF signaling responsible for neuritogenesis in early postnatal spiral ganglion.


Brain Research | 2012

Ras/p38 and PI3K/Akt but not Mek/Erk signaling mediate BDNF-induced neurite formation on neonatal cochlear spiral ganglion explants.

Lina Mullen; Kwang K. Pak; Eduardo Chavez; Kenji Kondo; Yves Brand; Allen F. Ryan

Neurotrophins participate in regulating the survival, differentiation, and target innervation of many neurons, mediated by high-affinity Trk and low-affinity p75 receptors. In the cochlea, spiral ganglion (SG) neuron survival is strongly dependent upon neurotrophic input, including brain-derived neurotrophic factor (BDNF), which increases the number of neurite outgrowth in neonatal rat SG in vitro. Less is known about signal transduction pathways linking the activation of neurotrophin receptors to SG neuron nuclei. In particular, the p38 and cJUN Kinase (JNK), mitogen-activated protein kinase (MAPK) pathways, which participate in JNK signaling in other neurons, have not been studied. We found that inhibition of Ras, p38, phosphatidyl inositol 3 kinase (PI3K) or Akt signaling reduced or eliminated BDNF mediated increase in number of neurite outgrowth, while inhibition of Mek/Erk had no influence. Inhibition of Rac/cdc42, which lies upstream of JNK, modestly enhanced BDNF induced formation of neurites. Western blotting implicated p38 and Akt signaling, but not Mek/Erk. The results suggest that the Ras/p38 and PI3K/Akt are the primary pathways by which BDNF promotes its effects. Activation of Rac/cdc42/JNK signaling by BDNF may reduce the formation of neurites. This is in contrast to our previous results on NT-3, in which Mek/Erk signaling was the primary mediator of SG neurite outgrowth in vitro. Our data on BDNF agree with prior results from others that have implicated PI3K/Akt involvement in mediating the effects of BDNF on SG neurons in vitro, including neuronal survival and neurite extension. However, the identification of p38 and JNK involvement is entirely novel. The results suggest that neurotrophins can exert opposing effects on SG neurons, the balance of competing signals influencing the generation of neurites. This competition could provide a potential mechanism for the control of neurite number during development.


BMC Neuroscience | 2011

Simvastatin protects auditory hair cells from gentamicin-induced toxicity and activates Akt signaling in vitro

Yves Brand; Cristian Setz; Soledad Levano; Alwin Listyo; Eduardo Chavez; Kwang Pak; Michael Sung; Vesna Radojevic; Allen F. Ryan; Daniel Bodmer

BackgroundInhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, known as statins, are commonly used as cholesterol-lowering drugs. During the past decade, evidence has emerged that statins also have neuroprotective effects. Research in the retina has shown that simvastatin, a commonly used statin, increases Akt phosphorylation in vivo, indicating that the PI3K/Akt pathway contributes to the protective effects achieved. While research about neuroprotective effects have been conducted in several systems, the effects of statins on the inner ear are largely unknown.ResultsWe evaluated whether the 3-hydroxy-3-methylglutaryl-coenzyme A reductase is present within the rat cochlea and whether simvastatin is able to protect auditory hair cells from gentamicin-induced apoptotic cell death in a in vitro mouse model. Furthermore, we evaluated whether simvastatin increases Akt phosphorylation in the organ of Corti. We detected 3-hydroxy-3-methylglutaryl-coenzyme A reductase mRNA in organ of Corti, spiral ganglion, and stria vascularis by reverse transcriptase-polymerase chain reaction (RT-PCR). Moreover, we observed a dose-dependent and significant reduction of hair cell loss in organs of Corti treated with simvastatin in addition to gentamicin, as compared to samples treated with gentamicin alone. The protective effect of simvastatin was reversed by addition of mevalonate, a downstream metabolite blocked by simvastatin, demonstrating the specificity of protection. Finally, Western blotting showed an increase in organ of Corti Akt phosphorylation after simvastatin treatment in vitro.ConclusionThese results suggest a neuroprotective effect of statins in the inner ear, mediated by reduced 3-hydroxy-3-methylglutaryl-coenzyme A reductase metabolism and Akt activation.


PLOS ONE | 2014

A Mouse Model of Otitis Media Identifies HB-EGF as a Mediator of Inflammation-Induced Mucosal Proliferation

Keigo Suzukawa; Julia Tomlin; Kwang Pak; Eduardo Chavez; Arwa Kurabi; Andrew Baird; Stephen I. Wasserman; Allen F. Ryan

Objective Otitis media is one of the most common pediatric infections. While it is usually treated without difficulty, up to 20% of children may progress to long-term complications that include hearing loss, impaired speech and language development, academic underachievement, and irreversible disease. Hyperplasia of middle ear mucosa contributes to the sequelae of acute otitis media and is of important clinical significance. Understanding the role of growth factors in the mediation of mucosal hyperplasia could lead to the development of new therapeutic interventions for this disease and its sequelae. Methods From a whole genome gene array analysis of mRNA expression during acute otitis media, we identified growth factors with expression kinetics temporally related to hyperplasia. We then tested these factors for their ability to stimulate mucosal epithelial growth in vitro, and determined protein levels and histological distribution in vivo for active factors. Results From the gene array, we identified seven candidate growth factors with upregulation of mRNA expression kinetics related to mucosal hyperplasia. Of the seven, only HB-EGF (heparin-binding-epidermal growth factor) induced significant mucosal epithelial hyperplasia in vitro. Subsequent quantification of HB-EGF protein expression in vivo via Western blot analysis confirmed that the protein is highly expressed from 6 hours to 24 hours after bacterial inoculation, while immunohistochemistry revealed production by middle ear epithelial cells and infiltrating lymphocytes. Conclusion Our data suggest an active role for HB-EGF in the hyperplasia of the middle ear mucosal epithelium during otitis media. These results imply that therapies targeting HB-EGF could ameliorate mucosal growth during otitis media, and thereby reduce detrimental sequelae of this childhood disease.


International Journal of Neuroscience | 2013

Changes in responsiveness of rat spiral ganglion neurons to neurotrophins across age: differential regulation of survival and neuritogenesis.

Kenji Kondo; Kwang Pak; Eduardo Chavez; Lina Mullen; Sara Euteneuer; Allen F. Ryan

Developmental changes in responsiveness of rat spiral ganglion neurons (SGNs) to neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF) were examined using an explant culture system. Spiral ganglion (SG) explants at embryonic Day 18 (E18), postnatal Day 0 (P0), P5, P10 and P20 were cultured with the addition of either NT-3 or BDNF at various concentrations (0.1–100 ng/ml) and analyzed the dose-response characteristics of three parameters: SGN survival, the number of neurites emanating from the explants and the length of neurite extension. In E18 cultures, SGN survival and neurite number were enhanced more strongly by NT-3 than by the BDNF. As the explants became more mature, the effects of NT-3 decreased, whereas those of BDNF increased, peaking at P0. Although the intrinsic capacity of SGNs to produce and extend neurites declined considerably by P20, they still retained the capacity to respond to both NT-3 and BDNF. These temporal patterns in responsiveness of SGNs to neurotrophins correspond well to the expression pattern of the two neurotrophins in cochlear sensory epithelium in vivo and also correlate with the time course of developmental events in SGNs such as cell death and the establishment of mature hair cell innervation patterns.


Journal of Molecular Neuroscience | 2013

Neural Cell Adhesion Molecule L1 Modulates Type I But Not Type II Inner Ear Spiral Ganglion Neurite Outgrowth in an In Vitro Alternate Choice Assay

Yves Brand; Michael Sung; Eduardo Chavez; Eric Wei; Kwang K. Pak; Gary D. Housley; Daniel Bodmer; Allen F. Ryan

L1, a neural cell adhesion molecule of the immunoglobulin superfamily, is widely expressed in the nervous system and important in axonal outgrowth, guidance, synapse formation, and signaling. Gene deletion studies emphasize the significance of L1 during development of the central nervous system and L1 is crucial for the topographic targeting of retinal axons. In contrast to the brain and retina, the role of L1 in the inner ear is largely unknown. While previous studies have localized L1 in the developing inner ear of the chicken and mouse, its function during the innervation of the cochlea still remains largely unclear. We therefore investigated the functional role of L1 in the mammalian inner ear. Our aim was to determine whether or not L1 can modulate type I and/or type II spiral ganglion neuron outgrowth using an in vitro alternate choice assay. We found that L1, presented in stripe micropatterns, provide directional cues to neonatal rodent type I but not type II inner ear spiral ganglion neurites. The results suggest that L1 may play a role in axonal pathfinding of type I spiral ganglion dendrites toward their inner hair cell targets but not of type II toward the outer hair cells.


Journal of Molecular Neuroscience | 2015

Neural Cell Adhesion Molecule NrCAM Is Expressed in the Mammalian Inner Ear and Modulates Spiral Ganglion Neurite Outgrowth in an In Vitro Alternate Choice Assay

Yves Brand; Michael Sung; Kwang Pak; Eduardo Chavez; Eric Wei; Vesna Radojevic; Daniel Bodmer; Allen F. Ryan

Neuron-glial-related cell adhesion molecule (NrCAM) is a neuronal cell adhesion molecule involved in neuron–neuron and neuron–glial adhesion as well as directional signaling during axonal cone growth. NrCAM has been shown to be involved in several cellular processes in the central and peripheral nervous systems, including neurite outgrowth, axonal pathfinding and myelination, fasciculation of nerve fibers, and cell migration. This includes sensory systems such as the eye and olfactory system. However, there are no reports on the expression/function of NrCAM in the auditory system. The aim of the present study was to elucidate the occurrence of NrCAM in the mammalian cochlea and its role in innervation of the auditory end organ. Our work indicates that NrCAM is highly expressed in the developing mammalian cochlea (position consistent with innervation). Moreover, we found that NrCAM, presented in stripe micropatterns, provide directional cues to neonatal rat inner ear spiral ganglion neurites in vitro. Our results are consistent with a role for NrCAM in the pathfinding of spiral ganglion dendrites toward their hair cell targets in the sensory epithelium.


Otolaryngology-Head and Neck Surgery | 2008

R443 – Organotypic Co-Culture of Spiral Ganglion and Organ of Corti

Jacob Husseman; Kwang Pak; Eduardo Chavez; Allen F. Ryan

Problem The ability to carry out in vitro culture of the auditory neuroepithelium has provided a powerful means of studying inner ear development. Recently, we have developed an organotypic culture technique that mimics the perinatal cochlea in vivo. Methods Using sterile microdissection and in vitro methods, we have been able to co-culture explanted spiral ganglion (SG) with separate explanted organ of Corti (oC) from different neonatal mice. The SG and oC were co-cultured in their correct anatomical positions. Success of the technique appears dependent on the use of culture plate inserts which prevent cellular attachment to the plastic culture surface and the resulting migration of neurons and hair cells (HCs). Results Using this technique, we have noted an average of 5–10 neurites per SG explant growing into the oC via the spiral lamina, following the anatomic pathway used during development. Confocal microscopy was used to visualize the contact area between dendritic growth and HCs. This demonstrates branching of neurites to multiple inner HCs as well as some branches extending to outer HCs. This pattern is consistent with early cochlear development. In contrast, with alternate techniques, neurites followed very different paths to the oC, sometimes innervating inner hair cells from the outer hair cell side of the epithelium. Conclusion This culture system offers a unique capacity to evaluate factors affecting spiral ganglion neurite guidance via manipulation of either the oC or SG. Significance The use of separated SG and oC will allow tissue from knockout mice to be paired with wild-type tissue, to explore fundamental mechanisms involved in cochlear innervation. Clinical implications include the ability to optimize SG ingrowth to cochlear implant electrodes offering tremendous potential for improving frequency resolution and dynamic range, as well as the ability to direct SG dendrites to regenerated/transplanted HCs as such advances develop.


Developmental Neurobiology | 2007

Laminin and Fibronectin Modulate Inner Ear Spiral Ganglion Neurite Outgrowth in an In Vitro Alternate Choice Assay

Amaretta Evans; Sara Euteneuer; Eduardo Chavez; Lina Mullen; Elliot E. Hui; Sangeeta N. Bhatia; Allen F. Ryan

Collaboration


Dive into the Eduardo Chavez's collaboration.

Top Co-Authors

Avatar

Allen F. Ryan

University of California

View shared research outputs
Top Co-Authors

Avatar

Kwang Pak

University of California

View shared research outputs
Top Co-Authors

Avatar

Lina Mullen

University of California

View shared research outputs
Top Co-Authors

Avatar

Yves Brand

University of California

View shared research outputs
Top Co-Authors

Avatar

Daniel Bodmer

University of California

View shared research outputs
Top Co-Authors

Avatar

Michael Sung

University of California

View shared research outputs
Top Co-Authors

Avatar

Sara Euteneuer

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Wei

University of California

View shared research outputs
Top Co-Authors

Avatar

Kwang K. Pak

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge