Network


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

Hotspot


Dive into the research topics where Lina Mullen is active.

Publication


Featured researches published by Lina Mullen.


Jaro-journal of The Association for Research in Otolaryngology | 2001

Ras/MEK but not p38 signaling mediates NT-3-induced neurite extension from spiral ganglion neurons.

Christoph Aletsee; Andrew Beros; Lina Mullen; Sean D. Palacios; Kwang Pak; Stefan Dazert; Allen F. Ryan

Neurotrophin (NT)-3 is expressed in the neuronal target tissue of the developing rat cochlea and has been shown to promote the survival and neurite outgrowth of spiral ganglion (SG) neurons, suggesting a role for this protein during the innervation of the organ of Corti. In other neurons, NT-3 can mediate neuritogenesis and survival via a number of intracellular signal pathways. To date, the intracellular transduction pathways involved in the mediation of NT-3 effects have not been investigated in SG neurons. To determine whether the activities of NT-3 on SG neurons are dependent on the activation of mitogen-activated protein kinase kinases (MEK)/extracellular-signal-regulated kinases (ERK), Ras, and/or p38, SG explants from postnatal-day 4 rats were cultured with NT-3 and increasing concentrations of the MEK inhibitor U0126, the Ras farnesyl-transferase inhibitor (FTI)-277, and the p38 inhibitor SB203580. After fixation and immunocytochemical labeling, neurite growth was evaluated. A dose-dependent decrease of the effects of NT-3 on length and number of processes was observed in the U0126- and FTI-277-treated SG neurons. In contrast, SB203580 had no significant effect on NT-3-mediated stimulation of neurite growth, in terms of either number or length. The results suggest that NT-3 effects on SG neurons are mediated primarily by the Ras/MEK/ERK signaling pathway.


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.


Audiology and Neuro-otology | 2001

The Disintegrin Kistrin Inhibits Neurite Extension from Spiral Ganglion Explants Cultured on Laminin

Christoph Aletsee; Lina Mullen; David W. Kim; Kwang Pak; Dominik Brors; Stefan Dazert; Allen F. Ryan

The influence of laminin-1 (LN) and tenascin-C (TN), extracellular matrix molecules expressed spatially and temporally along the neural growth route from spiral ganglion (SG) neurons to the cochlear sensory cells, was evaluated in cultured SG explants from postnatal day 4 rats. Increasing concentrations of LN resulted in a strong, dose-dependent increase in the length of neurites and in a higher number of neural processes, while varying TN concentrations had relatively minor effects on both parameters. The results suggest differential receptor activation by LN and TN. When explants grown on LN were treated with Kistrin, an inhibitor of the αvβ3 integrin, the LN-induced increase in neurite length was reduced in a dose-dependent manner. However, the number of extending neurites was not affected, indicating that different receptors mediate this response, perhaps by increasing neuronal survival.


Journal of Neuroimmunology | 2002

Inflammatory signals increase Fas ligand expression by inner ear cells

Daniel Bodmer; Dominik Brors; Kwang Pak; Elizabeth M. Keithley; Lina Mullen; Allen F. Ryan; Bertrand Gloddek

There is considerable evidence that hearing and vestibular function can be influenced by immune processes. The inner ear has evolved mechanisms, such as the blood-labyrinthine barrier that limit immune responses and autoimmune processes to reduce the potential for damage to cochlear cells. Recently, expression of Fas ligand (FasL) in some non-lymphoid tissue, as in the anterior chamber of the eye, has been hypothesized to play a role in protection of sensitive organs from activated T-cells. We show that under resting conditions, cochlear cells express little or no FasL. However, after exposure to interferon-gamma in vitro, FasL is induced in many neonatal cochlear cells. In addition, we show that FasL is upregulated in adult cochlear cells after induction of a sterile labyrinthitis in vivo. The induction of FasL by inflammation may serve to limit cochlear immune responses, and to protect sensorineural tissue from immune and autoimmune damage.


Hearing Research | 2002

The effects of laminin-1 on spiral ganglion neurons are dependent on the MEK/ERK signaling pathway and are partially independent of Ras.

Christoph Aletsee; Dominik Brors; Sean D. Palacios; Kwang Pak; Lina Mullen; Stefan Dazert; Allen F. Ryan

Laminin-1 (LN) is expressed along the route of neural growth from spiral ganglion (SG) neurons towards the developing organ of Corti, and has been shown to enhance neurite outgrowth from SG neurons in vitro. Signal transduction pathways linking LN signaling at the cell membrane to the cell nucleus can involve a variety of signaling molecules. Data from other systems suggest the potential involvement of the small G protein Ras, and the mitogen-activated protein kinases (MAPKs) Erk and/or p38. To assess these possibilities, the length and number of processes extending from SG explants cultured on LN-coated surfaces were evaluated after treatment with the Ras inhibitor FTI-277, the p38 inhibitor SB203580 and MAPK kinase (MEK) inhibitor U0126, which operates immediately upstream of the Erk MAPK. Treatment with the Ras inhibitor at levels known to inhibit the H- and N-Ras isoforms had no effect, while FTI-277 levels known to inhibit K-Ras reduced only neurite length. Suppression of MEK resulted in a decrease of both parameters, while incubation with the p38 inhibitor had no effect. The results of this study suggest that MEK plays a central role in LN signaling in SG neurites. While K-Ras signaling may participate in MEK-dependent increases in neurite length, the MEK-dependent increase in neurite number appears to be activated by a different intracellular pathway.


Audiology and Neuro-otology | 2006

Environmental Micropatterning for the Study of Spiral Ganglion Neurite Guidance

Allen F. Ryan; John H. Wittig; Amaretta Evans; Stefan Dazert; Lina Mullen

The projection of neuronal processes is guided by a variety of soluble and insoluble factors, which are sensed by a fiber’s growth cone. It is the differential distribution of such guidance cues that determine the direction in which neurites grow. The growth cone senses these cues on a fine scale, using extensible filopodia that range from a few to tens of µm in length. In order to study the effects of guidance cues on spiral ganglion (SG) neurites, we have used methods for distributing both soluble and insoluble cues on a scale appropriate for sensing by growth filopodia. The scale of these methods are at the micro, rather than nano, level to match the sensing range of the growth cone. Microfluidics and transfected cells were used to spatially localize tropic factors within the fluid environment of extending neurites. Micropatterning was used to present neurites with stripes of insoluble factors. The results indicate that differentially distributed permissive, repulsive and stop signals can control the projection of SG neurites. Implications for future micropatterning studies, for SG development and for the growth of deafferented SG dendrites toward a cochlear implant are discussed.


Advances in oto-rhino-laryngology | 2002

Immunological damage to the inner ear: current and future therapeutic strategies.

Allen F. Ryan; Kwang Pak; W. Low; A. Battaglia; Lina Mullen; Jeffrey P. Harris; Elizabeth M. Keithley

There is considerable evidence to suggest that hearing and vestibular function can be influenced by immunity in the inner ear. While immunity can protect against infections of the labyrinth, immune response also has the capacity to damage the delicate tissues of the inner ear. Antigenic challenge of the inner ear of sensitized animals leads to rapid accumulation of leukocytes, antibody production, hearing loss and tissue damage. Moreover, a number of systemic autoimmune disorders include hearing loss and vertigo as part of their constellation of symptoms. It also appears that autoimmune damage can exist as an entity confined to the labyrinth. Immune disorders of the inner ear are of special interest since they are among the few forms of hearing loss that are currently amenable to medical treatment. In addition, recent developments in understanding the intracellular pathways that participate in damage to the inner ear provide new opportunities for pharmacotherapy of immune-mediated disorders of hearing and balance.


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.


Advances in oto-rhino-laryngology | 2009

Cellular Targeting for Cochlear Gene Therapy

Allen F. Ryan; Lina Mullen; Joni K. Doherty

Gene therapy has considerable potential for the treatment of disorders of the inner ear. Many forms of inherited hearing loss have now been linked to specific locations in the genome, and for many of these the genes and specific mutations involved have been identified. This information provides the basis for therapy based on genetic approaches. However, a major obstacle to gene therapy is the targeting of therapy to the cells and the times that are required. The inner ear is a very complex organ, involving dozens of cell types that must function in a coordinated manner to result in the formation of the ear, and in hearing. Mutations that result in hearing loss can affect virtually any of these cells. Moreover, the genes involved are active during particular times, some for only brief periods of time. In order to be effective, gene therapy must be delivered to the appropriate cells, and at the appropriate times. In many cases, it must also be restricted to these cells and times. This requires methods with which to target gene therapy in space and time. Cell-specific gene promoters offer the opportunity to direct gene therapy to a desired cell type. Moreover, conditional promoters allow gene expression to be turned off and on at desired times. Theoretically, these technologies offer a mechanism by which to deliver gene therapy to any cell, at any given time. This chapter will examine the potential for such targeting to deliver gene therapy to the inner ear in a precisely controlled manner.


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 Lina Mullen'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

Eduardo Chavez

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amaretta Evans

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Li

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sara Euteneuer

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge