Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
World Neurosurg ; 116: e525-e533, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29772365

ABSTRACT

OBJECTIVE: Approximately 12% of intracerebral hemorrhages (ICHs) occur in the thalamus. Understanding the anatomic regions involved with thalamic hemorrhages is potentially useful, offering the physician a more accurate prognosis for patient outcomes. This study was performed to determine if thalamic hemorrhage location observed on a computed tomography (CT) scan was predictive of neurologic outcomes. METHODS: A sample of 168 thalamic hemorrhage patients admitted to a tertiary care center were analyzed. Axial CT scans of thalamic hemorrhages were classified into 1 of 6 possible categories based on thalamic nuclei anatomy: anterior, posterior, medial, lateral, central, or global. For each classification, patient clinical characteristics were collected to identify variables indicative of clinical outcome. Outcome measures used in this study included mortality, hospital length of stay, readmission within 30 days, ICH score, Glasgow Coma Scale score, neurologic deterioration (calculated as a change in modified Rankin scale score from admission to discharge), and discharge disposition. RESULTS: On multivariable analysis, patients with posterior and lateral thalamic hemorrhages demonstrated a decreased likelihood of mortality; patients with posterior hemorrhages were less likely to have neurologic deterioration relative to global thalamic hemorrhages when controlling for hemorrhage volume and ventriculomegaly. Ventriculomegaly and hemorrhage volume were also predictive of both mortality and neurologic deterioration. CONCLUSIONS: In thalamic hemorrhages, patient prognosis may be influenced by hemorrhage location, with posterior and lateral hemorrhages demonstrating better clinical outcome versus hemorrhages in other locations. This is potentially valuable because hemorrhage location affords the treating physician a readily available prognostic factor when assessing intracranial hemorrhages.


Subject(s)
Cerebral Hemorrhage/diagnostic imaging , Length of Stay/trends , Tertiary Care Centers/trends , Thalamus/diagnostic imaging , Aged , Cerebral Hemorrhage/mortality , Cerebral Hemorrhage/surgery , Female , Humans , Male , Middle Aged , Mortality/trends , Prognosis , Retrospective Studies , Thalamus/surgery , Tomography, X-Ray Computed/methods
2.
PLoS One ; 11(6): e0157768, 2016.
Article in English | MEDLINE | ID: mdl-27351484

ABSTRACT

Unlike mammals, the non-mammalian vertebrate inner ear can regenerate the sensory cells, hair cells, either spontaneously or through induction after hair cell loss, leading to hearing recovery. The mechanisms underlying the regeneration are poorly understood. By microarray analysis on a chick model, we show that chick hair cell regeneration involves the activation of proliferation genes and downregulation of differentiation genes. Both MYC and FGF are activated in chick hair cell regeneration. Using a zebrafish lateral line neuromast hair cell regeneration model, we show that the specific inhibition of Myc or Fgf suppresses hair cell regeneration, demonstrating that both pathways are essential to the process. Rapid upregulation of Myc and delayed Fgf activation during regeneration suggest a role of Myc in proliferation and Fgf in differentiation. The dorsal-ventral pattern of fgfr1a in the neuromasts overlaps with the distribution of hair cell precursors. By laser ablation, we show that the fgfr1a-positive supporting cells are likely the hair cell precursors that directly give rise to new hair cells; whereas the anterior-posterior fgfr1a-negative supporting cells have heightened proliferation capacity, likely to serve as more primitive progenitor cells to replenish lost precursors after hair cell loss. Thus fgfr1a is likely to mark compartmentalized supporting cell subtypes with different capacities in renewal proliferation and hair cell regeneration. Manipulation of c-MYC and FGF pathways could be explored for mammalian hair cell regeneration.


Subject(s)
Fibroblast Growth Factors/metabolism , Lateral Line System/metabolism , Mast Cells/metabolism , Neurons, Afferent/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Regeneration , Animals , Cell Proliferation , Fibroblast Growth Factors/genetics , Lateral Line System/cytology , Lateral Line System/physiology , Mast Cells/cytology , Mast Cells/physiology , Neurons, Afferent/cytology , Neurons, Afferent/physiology , Proto-Oncogene Proteins c-myc/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
PLoS One ; 10(6): e0130827, 2015.
Article in English | MEDLINE | ID: mdl-26121033

ABSTRACT

Noise-induced hearing loss (NIHL) is the most significant occupational health issue worldwide. We conducted a genome-wide association study to identify single-nucleotide polymorphisms (SNPs) associated with hearing threshold shift in young males undergoing their first encounter with occupational impulse noise. We report a significant association of SNP rs7598759 (p < 5 x 10(-7); p = 0.01 after permutation and correction; Odds Ratio = 12.75) in the gene coding for nucleolin, a multifunctional phosphoprotein involved in the control of senescence and protection against apoptosis. Interestingly, nucleolin has been shown to mediate the anti-apoptotic effect of HSP70, a protein found to prevent ototoxicity and whose polymorphisms have been associated with susceptibility to NIHL. Increase in nucleolin expression has also been associated with the prevention of apoptosis in cells undergoing oxidative stress, a well-known metabolic sequela of noise exposure. To assess the potential role of nucleolin in hearing loss, we tested down-regulation of nucleolin in cochlear sensory cells HEI-OC1 under oxidative stress conditions and report increased sensitivity to cisplatin, a chemotherapeutic drug with ototoxic side effects. Additional SNPs were found with suggestive association (p < 5 x 10(-4)), of which 7 SNPs were located in genes previously reported to be related to NIHL and 43 of them were observed in 36 other genes previously not reported to be associated with NIHL. Taken together, our GWAS data and in vitro studies reported herein suggest that nucleolin is a potential candidate associated with NIHL in this population.


Subject(s)
Auditory Threshold , Genome-Wide Association Study , Hearing/genetics , Noise, Occupational , Polymorphism, Single Nucleotide/genetics , Audiometry , Cell Nucleus/metabolism , Cell Survival , Down-Regulation/genetics , Genetic Association Studies , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/physiopathology , Humans , Linkage Disequilibrium/genetics , Male , Oxidative Stress , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Nucleolin
4.
PLoS One ; 8(2): e55359, 2013.
Article in English | MEDLINE | ID: mdl-23383324

ABSTRACT

Inner ear sensory hair cells die following exposure to aminoglycoside antibiotics or chemotherapeutics like cisplatin, leading to permanent auditory and/or balance deficits in humans. Zebrafish (Danio rerio) are used to study drug-induced sensory hair cell death since their hair cells are similar in structure and function to those found in humans. We developed a cisplatin dose-response curve using a transgenic line of zebrafish that expresses membrane-targeted green fluorescent protein under the control of the Brn3c promoter/enhancer. Recently, several small molecule screens have been conducted using zebrafish to identify potential pharmacological agents that could be used to protect sensory hair cells in the presence of ototoxic drugs. Dimethyl sulfoxide (DMSO) is typically used as a solvent for many pharmacological agents in sensory hair cell cytotoxicity assays. Serendipitously, we found that DMSO potentiated the effects of cisplatin and killed more sensory hair cells than treatment with cisplatin alone. Yet, DMSO alone did not kill hair cells. We did not observe the synergistic effects of DMSO with the ototoxic aminoglycoside antibiotic neomycin. Cisplatin treatment with other commonly used organic solvents (i.e. ethanol, methanol, and polyethylene glycol 400) also did not result in increased cell death compared to cisplatin treatment alone. Thus, caution should be exercised when interpreting data generated from small molecule screens since many compounds are dissolved in DMSO.


Subject(s)
Cell Death/drug effects , Cisplatin/pharmacology , Dimethyl Sulfoxide/pharmacology , Hair Cells, Ampulla/drug effects , Analysis of Variance , Animals , Animals, Genetically Modified , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Synergism , Green Fluorescent Proteins/metabolism , Mass Spectrometry , Microscopy, Confocal , Xanthenes , Zebrafish
5.
Hear Res ; 298: 93-103, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23296212

ABSTRACT

In animals, hearing loss resulting from cochlear mechanosensory cell damage can be mitigated by antioxidants such as d-methionine (d-met) and acetyl-l-carnitine (ALCAR). The systemic routes of administration of these compounds, that must of necessity transit trough the cochlear fluids, may affect the antioxidant levels in the cochlea and the resulting oto-protective effect. In this study, we analyzed the pharmacokinetics of [(14)C]d-met in the cochlea and four other tissues after intratracheal (IT), intranasal (IN), and oral by gavage (OG) administration and compared it to intravenous administration (IV). We then analyzed the effect of these four routes on the antioxidant content of the cochlear fluids after d-met or ALCAR administration, by liquid chromatography/mass spectrometry. Our results showed that the concentration of methionine and ALCAR in cochlear fluids significantly increased after their respective systemic administration. Interestingly, d-met administration also contributed to an increase of ALCAR. Our results also showed that the delivery routes differently affected the bioavailability of administered [(14)C]d-met as well as the concentrations of methionine, ALCAR and the ratio of oxidized to reduced glutathione. Overall, pulmonary delivery via IT administration achieved high concentrations of methionine, ALCAR, and oxidative-related metabolites in cochlear fluids, in some cases surpassing IV administration, while IN route appeared to be the least efficacious. To our knowledge, this is the first report of the direct measurements of antioxidant levels in cochlear fluids after their systemic administration. This report also demonstrates the validity of the pulmonary administration of antioxidants and highlights the different contributions of d-met and ALCAR allowing to further investigate their impact on oxidative stress in the cochlear microenvironment.


Subject(s)
Acetylcarnitine/administration & dosage , Acetylcarnitine/pharmacokinetics , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Glutathione/metabolism , Labyrinthine Fluids/metabolism , Methionine/administration & dosage , Methionine/pharmacokinetics , Administration, Inhalation , Administration, Intranasal , Administration, Oral , Animals , Biological Availability , Biotransformation , Chromatography, High Pressure Liquid , Endolymph/metabolism , Injections, Intravenous , Male , Mass Spectrometry , Oxidation-Reduction , Oxidative Stress/drug effects , Perilymph/metabolism , Rats , Rats, Sprague-Dawley
6.
J Histochem Cytochem ; 59(12): 1060-75, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22147606

ABSTRACT

Identification of cell types in tumor-associated stroma that are involved in the development of melanoma is hampered by their heterogeneity. The authors used flow cytometry and immunohistochemistry to demonstrate that anti-MART-1 antibodies can discriminate between melanoma and stroma cells. They investigated the cellular composition of the MART-1-, non-hematopoietic melanoma-associated stroma, finding it consisted mainly of Sca-1+ and CD146+ cells. These cell types were also observed in the skin and muscle adjacent to developing melanomas. The Sca-1+ cell population was observed distributed in the epidermis, hair follicle bulges, and tumor capsule. The CD146+ population was found distributed within the tumor, mainly associated with blood vessels in a perivascular location. In addition to a perivascular distribution, CD146+ cells expressed α-smooth muscle actin, lacked expression of endothelial markers CD31 and CD34, and were therefore identified as pericytes. Pericytes were found to be associated with CD31+ endothelial cells; however, some pericytes were also observed associated with CD31-, MART-1+ B16 melanoma cells that appeared to form blood vessel structures. Furthermore, the authors observed extensive nuclear expression of HIF-1α in melanoma and stroma cells, suggesting hypoxia is an important factor associated with the melanoma microenvironment and vascularization. The results suggest that pericytes and Sca-1+ stroma cells are important contributors to melanoma development.


Subject(s)
Antigens, Ly/metabolism , MART-1 Antigen/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/metabolism , Pericytes/pathology , Animals , CD146 Antigen/metabolism , Cell Hypoxia , Cell Line, Tumor , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred ICR , Mice, SCID , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pericytes/metabolism , Skin/metabolism , Skin/pathology , Stromal Cells/metabolism , Time Factors , Tumor Microenvironment
7.
Hear Res ; 266(1-2): 18-25, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20438823

ABSTRACT

The discovery of avian cochlear hair cell regeneration in the late 1980s and the concurrent development of new techniques in molecular and developmental biology generated a renewed interest in understanding the genetic mechanisms that regulate hair cell development in the embryonic avian and mammalian cochlea and regeneration in the mature avian cochlea. Research from many labs has demonstrated that the development of the inner ear utilizes a complex series of genetic signals and pathways to generate the endorgans, specify cell identities, and establish innervation patterns found in the inner ear. Recent studies have shown that the Notch signaling pathway, the Atoh1/Hes signaling cascade, the stem cell marker Sox2, and some of the unconventional myosin motor proteins are utilized to regulate distinct steps in inner ear development. While many of the individual genes involved in these pathways have been identified from studies of mutant and knockout mouse cochleae, the interplay of all these signals into a single systemic program that directs this process needs to be explored. We need to know not only what genes are involved, but understand how their gene products interact with one another in a structural and temporal framework to guide hair cell and supporting cell differentiation and maturation.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Cochlea/physiology , Hair Cells, Auditory/physiology , Regeneration , Animals , Birds , Cell Differentiation/genetics , Cell Lineage/genetics , Cochlea/cytology , Cochlea/growth & development , Gene Expression Regulation, Developmental , Humans , Mice , Regeneration/genetics , Signal Transduction
8.
Laryngoscope ; 119(9): 1770-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19554638

ABSTRACT

OBJECTIVES/HYPOTHESIS: The avian cochlea regenerates hair cells following aminoglycoside treatment through supporting cell proliferation. Immunocytochemical labeling of 5-bromo-2'-deoxyuridine (BrdU), a thymidine analog, is a popular nonradioactive marker for identifying cells in the DNA synthesis (S phase) of the cell cycle. However, it requires harsh treatments to denature double-stranded DNA for the antibody to bind BrdU. We explored a new method using 5-ethynyl-2'-deoxyuridine (EdU) as a thymidine analog and a nonantibody azide/alkyne reaction between EdU and the fluorescent probe. We propose that EdU is as effective as BrdU, but without the requirement for harsh denaturation or the use of antibodies for detection. STUDY DESIGN: Two-week-old chicks received a single gentamicin injection followed by a single EdU injection 72 hours later. Cochleae were extracted 4-8 hours later, fixed, and processed for fluorescent detection of EdU. METHODS: Cochleae were processed for detection of incorporated EdU using the Click-iT Imaging Kit (Invitrogen/Molecular Probes, Carlsbad, CA) and colabeled with Sox2, myosin VI, or myosin VIIa antibodies. Whole-mount cochlear preparations were examined with confocal microscopy. RESULTS: Supporting cells incorporated EdU into their newly synthesized DNA during the 4-8 hours following the EdU injection and were readily detected with little background signal. The intensity and quantity of cells labeled were similar to or better than that seen for BrdU. CONCLUSIONS: The EdU method is as effective as BrdU, without requiring harsh denaturation or secondary antibodies to identify proliferating cells. Thus, the nonantibody EdU system allows more flexibility by enabling colabeling with multiple antibodies to other cellular proteins involved in regeneration.


Subject(s)
Cochlea/cytology , Cochlea/physiology , Deoxyuridine/analogs & derivatives , Regeneration/physiology , Animals , Cell Proliferation , Chickens , Deoxyuridine/metabolism , Flow Cytometry/methods , Microscopy, Confocal , S Phase/physiology
9.
Hear Res ; 240(1-2): 1-11, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18487027

ABSTRACT

Aminoglycoside antibiotics induce caspase-dependent apoptotic death in cochlear hair cells. Apoptosis, a regulated form of cell death, can be induced by many stressors, which activate signaling pathways that result in the controlled dismantling of the affected cell. The caspase family of proteases is activated in the apoptotic signaling pathway and is responsible for cellular destruction. The initiator caspase-9 and the effector caspase-3 are both activated in chick cochlear hair cells following aminoglycoside exposure. We have analyzed caspase activation in the avian cochlea during gentamicin-induced hair cell death to compare two different methods of caspase detection: caspase antibodies and CaspaTag kits. Caspase antibodies bind to the cleaved activated form of caspase-9 or caspase-3 in specific locations in fixed tissue. CaspaTag is a fluorescent inhibitor that binds to a reactive cysteine residue on the large subunit of the caspase heterodimer in unfixed tissue. To induce cochlear hair cell loss, 1-2 week-old chickens received a single injection of gentamicin (300 mg/kg). Chicks were sacrificed 24, 30, 42, 48, 72, or 96 h after injection. Cochleae were dissected and labeled for activated caspase-9 or caspase-3 using either caspase-directed antibodies or CaspaTag kits. Ears were co-labeled with either phalloidin or myosin VI to visualize hair cells and to determine the progression of cochlear damage. The timing of caspase activation was similar for both assays; however, caspase-9 and caspase-3 antibodies labeled only those cells currently undergoing apoptotic cell death. Conversely, CaspaTag-labeled all the cells that have undergone apoptotic cell death and ejection from the sensory epithelium, in addition to those that are currently in the cell death process. This makes CaspaTag ideal for showing an overall pattern or level of cell death over a period of time, while caspase antibodies provide a snapshot of cell death at a specific time point.


Subject(s)
Anti-Bacterial Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cochlea/drug effects , Fluorescent Antibody Technique, Indirect , Gentamicins/toxicity , Reagent Kits, Diagnostic , Animals , Caspase Inhibitors , Chickens , Cochlea/enzymology , Cochlea/pathology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Fluorescent Dyes/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Microscopy, Fluorescence , Time Factors
10.
J Commun Disord ; 41(5): 421-43, 2008.
Article in English | MEDLINE | ID: mdl-18455177

ABSTRACT

UNLABELLED: Twenty years ago it was first demonstrated that birds could regenerate their cochlear hair cells following noise damage or aminoglycoside treatment. An understanding of how this structural and functional regeneration occurred might lead to the development of therapies for treatment of sensorineural hearing loss in humans. Recent experiments have demonstrated that noise exposure and aminoglycoside treatment lead to apoptosis of the hair cells. In birds, this programmed cell death induces the adjacent supporting cells to undergo regeneration to replace the lost hair cells. Although hair cells in the mammalian cochlea undergo apoptosis in response to noise damage and ototoxic drug treatment, the supporting cells do not possess the ability to undergo regeneration. However, current experiments on genetic manipulation, gene therapy, and stem cell transplantation suggest that regeneration in the mammalian cochlea may eventually be possible and may 1 day provide a therapeutic tool for hearing loss in humans. LEARNING OUTCOMES: The reader should be able to: (1) Describe the anatomy of the avian and mammalian cochlea, identify the individual cell types in the organ of Corti, and distinguish major features that participate in hearing function, (2) Demonstrate a knowledge of how sound damage and aminoglycoside poisoning induce apoptosis of hair cells in the cochlea, (3) Define how hair cell loss in the avian cochlea leads to regeneration of new hair cells and distinguish this from the mammalian cochlea where there is no regeneration following damage, and (4) Interpret the potential for new approaches, such as genetic manipulation, gene therapy and stem cell transplantation, could provide a therapeutic approach to hair cell loss in the mammalian cochlea.


Subject(s)
Genetic Therapy , Hearing Loss, Sensorineural/therapy , Aminoglycosides/toxicity , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Birds , Cell Proliferation/drug effects , Guinea Pigs , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/therapy , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Humans , Mice , Mice, Knockout , Microscopy, Confocal , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Organ of Corti/drug effects , Organ of Corti/pathology , Organ of Corti/physiopathology , Stem Cell Transplantation
11.
Biomaterials ; 29(8): 1028-42, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18037163

ABSTRACT

The overall goal of this work is to improve the culture of the auditory organ of birds for the dual use of developing a hair cell regeneration model and charting a pathway to the eventual replacement of the hearing organ. In doing so, we develop a protocol for removing the auditory organ from its basement membrane in the inner ear, attach the organ to a series of artificial basement membranes, and conduct qualitative and quantitative analysis of how cell morphology, viability and function change with time. Native matrix cultures, where the epithelium was floating in media with the basement membrane and accessory structures attached, were used as a basis of comparison. PuraMatrix, collagen I, collagen I/chondroitin-sulfate and Matrigel were chosen to encompass a diverse range of mechanical properties and macromolecule moieties. Surprisingly, we find that PuraMatrix outperformed the other matrices as a scaffold for sensory organ culture. PuraMatrix a self-assembled peptide hydrogel, is a biochemically specific culture substrate that contains none of the extracellular matrix (ECM) molecules and growth factors contained in the inner ear's basement membrane. Rheological measurements reveal that PuraMatrix may be a closer approximation to the stiffness of the soft tissue supporting the auditory organ. Cell density on the PuraMatrix substrate is comparable to that of the native matrix cultures, despite the absence of the basement membrane and accessory structures. Further studies show that PuraMatrix supports the culture of functional hair cells over a 72 h period, with a significant increase in the number of functional hair cells in comparison to the organ cultured without a matrix. This is the first example of adhesion of the adult auditory epithelium to a biomaterial for an extended period of time. With further optimization, this system will enable the performance of many novel biophysical and pharmacological studies involving hair cells and supporting cells.


Subject(s)
Cochlea/growth & development , Collagen/chemistry , Hydrogels/chemistry , Peptides/chemistry , Animals , Biocompatible Materials/chemistry , Cell Adhesion , Cell Count , Cell Survival , Chickens , Chondroitin Sulfates/chemistry , Cochlea/cytology , Drug Combinations , Elasticity , Epithelial Cells/cytology , Epithelium/growth & development , Extracellular Matrix/chemistry , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/cytology , Laminin/chemistry , Organ Culture Techniques/methods , Proteoglycans/chemistry , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism
12.
Int J Dev Biol ; 51(6-7): 633-47, 2007.
Article in English | MEDLINE | ID: mdl-17891722

ABSTRACT

Regeneration of sensory hair cells in the mature avian inner ear was first described just over 20 years ago. Since then, it has been shown that many other non-mammalian species either continually produce new hair cells or regenerate them in response to trauma. However, mammals exhibit limited hair cell regeneration, particularly in the auditory epithelium. In birds and other non-mammals, regenerated hair cells arise from adjacent non-sensory (supporting) cells. Hair cell regeneration was initially described as a proliferative response whereby supporting cells re-enter the mitotic cycle, forming daughter cells that differentiate into either hair cells or supporting cells and thereby restore cytoarchitecture and function in the sensory epithelium. However, further analyses of the avian auditory epithelium (and amphibian vestibular epithelium) revealed a second regenerative mechanism, direct transdifferentiation, during which supporting cells change their gene expression and convert into hair cells without dividing. In the chicken auditory epithelium, these two distinct mechanisms show unique spatial and temporal patterns, suggesting they are differentially regulated. Current efforts are aimed at identifying signals that maintain supporting cells in a quiescent state or direct them to undergo direct transdifferentiation or cell division. Here, we review current knowledge about supporting cell properties and discuss candidate signaling molecules for regulating supporting cell behavior, in quiescence and after damage. While significant advances have been made in understanding regeneration in non-mammals over the last 20 years, we have yet to determine why the mammalian auditory epithelium lacks the ability to regenerate hair cells spontaneously and whether it is even capable of significant regeneration under additional circumstances. The continued study of mechanisms controlling regeneration in the avian auditory epithelium may lead to strategies for inducing significant and functional regeneration in mammals.


Subject(s)
Epithelial Cells/physiology , Hair Cells, Auditory/physiology , Regeneration , Animals , Apoptosis/physiology , Birds/metabolism , Birds/physiology , Cell Cycle , Cell Differentiation/physiology , Cell Proliferation , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/cytology , Models, Biological , Receptors, Notch/metabolism , Signal Transduction
13.
Hear Res ; 232(1-2): 29-43, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17659854

ABSTRACT

Most cases of hearing loss are caused by the death or dysfunction of one of the many cochlear cell types. We examined whether cells from a neural stem cell line could replace cochlear cell types lost after exposure to intense noise. For this purpose, we transplanted a clonal stem cell line into the scala tympani of sound damaged mice and guinea pigs. Utilizing morphological, protein expression and genetic criteria, stem cells were found with characteristics of both neural tissues (satellite, spiral ganglion, and Schwann cells) and cells of the organ of Corti (hair cells, supporting cells). Additionally, noise-exposed, stem cell-injected animals exhibited a small but significant increase in the number of satellite cells and Type I spiral ganglion neurons compared to non-injected noise-exposed animals. These results indicate that cells of this neural stem cell line migrate from the scala tympani to Rosenthal's canal and the organ of Corti. Moreover, they suggest that cells of this neural stem cell line may derive some information needed from the microenvironment of the cochlea to differentiate into replacement cells in the cochlea.


Subject(s)
Cell Differentiation , Cell Movement , Cochlea/surgery , Hearing Loss, Noise-Induced/surgery , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Cell Death , Cell Line , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Female , Guinea Pigs , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/pathology , Labyrinth Supporting Cells/metabolism , Male , Mice , Neurons/transplantation , Spiral Ganglion/metabolism
14.
J Comp Neurol ; 499(5): 691-701, 2006 Dec 10.
Article in English | MEDLINE | ID: mdl-17048225

ABSTRACT

Hair cells of the inner ear are damaged by intense noise, aging, and aminoglycoside antibiotics. Gentamicin causes oxidative damage to hair cells, inducing apoptosis. In mammals, hair cell loss results in a permanent deficit in hearing and balance. In contrast, avians can regenerate lost hair cells to restore auditory and vestibular function. This study examined the changes of myosin VI and myosin VIIa, two unconventional myosins that are critical for normal hair cell formation and function, during hair cell death and regeneration. During the late stages of apoptosis, damaged hair cells are ejected from the sensory epithelium. There was a 4-5-fold increase in the labeling intensity of both myosins and a redistribution of myosin VI into the stereocilia bundle, concurrent with ejection. Two separate mechanisms were observed during hair cell regeneration. Proliferating supporting cells began DNA synthesis 60 hours after gentamicin treatment and peaked at 72 hours postgentamicin treatment. Some of these mitotically produced cells began to differentiate into hair cells at 108 hours after gentamicin (36 hours after bromodeoxyuridine (BrdU) administration), as demonstrated by the colabeling of myosin VI and BrdU. Myosin VIIa was not expressed in the new hair cells until 120 hours after gentamicin. Moreover, a population of supporting cells expressed myosin VI at 78 hours after gentamicin treatment and myosin VIIa at 90 hours. These cells did not label for BrdU and differentiated far too early to be of mitotic origin, suggesting they arose by direct transdifferentiation of supporting cells into hair cells.


Subject(s)
Apoptosis/physiology , Dyneins/metabolism , Hair Cells, Auditory/physiology , Myosin Heavy Chains/metabolism , Myosins/metabolism , Regeneration , Animals , Anti-Bacterial Agents/pharmacology , Chick Embryo , Gentamicins/pharmacology , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Immunohistochemistry , Mitosis/physiology , Myosin VIIa
15.
Drug Discov Today ; 10(19): 1307-12, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16214675

ABSTRACT

Deafness affects more than 40 million people in the UK and the USA, and many more world-wide. The primary cause of hearing loss is damage to or death of the sensory receptor cells in the inner ear, the hair cells. Birds can readily regenerate their cochlear hair cells but the mammalian cochlea has shown no ability to regenerate after damage. Current research efforts are focusing on gene manipulation, gene therapy and stem cell transplantation for repairing or replacing damaged mammalian cochlear hair cells, which could lead to therapies for treating deafness in humans.


Subject(s)
Ear, Inner/physiology , Regeneration , Animals , Apoptosis , Cochlea/physiology , Genes, Retinoblastoma , Genetic Therapy , Hair Cells, Auditory/physiology , Humans , Stem Cell Transplantation
16.
Exp Neurol ; 194(2): 320-32, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15992799

ABSTRACT

Neural stem cells (NSCs) are the most primordial and least committed cells of the nervous system, the cells that exist before regional specification develops. Because immunocytochemically-detectable markers that are sufficiently specific and sensitive to define an NSC have not yet been fully defined, we have taken the strong view that, to be termed a "stem cell" in the nervous system--in contrast to a "progenitor" or "precursor" (whose lineage commitment is further restricted)--a single neuroectodermally-derived cell must fulfill an operational definition that is essentially similar to that used in hematopoiesis. In other words, it must possess the following functional properties: (1) "Multipotency", i.e., the ability to yield mature cells in all three fundamental neural lineages throughout the nervous system--neurons (of all subtypes), astrocytes (of all types), oligodendrocytes--in multiple regional and developmental contexts and in a region and developmental stage-appropriate manner. (2) The ability to populate a developing region and/or repopulate an ablated or degenerated region of the nervous system with appropriate cell types. (3) The ability to be serially transplanted. (4) "Self-renewal", i.e., the ability to produce daughter cells (including new NSCs) with identical properties and potential. Having identified a murine neural cell clone that fulfills this strict operational definition--in contrast to other studies that used less rigorous or non-operational criteria for defining an NSC (e.g., the "neurosphere" assay)--we then examined, by comparing gene expression profiles, the relationship such a cell might have to (a) a multipotent somatic stem cell from another organ system (the hematopoietic stem cell [HSC]); (b) a pluripotent stem cell derived from the inner cell mass and hence without organ assignment (an embryonic stem cell); (c) neural cells isolated and maintained primarily as neurospheres but without having been subjected to the above mentioned operational screen ("CNS-derived neurospheres"). ESCs, HSCs, and operationally-defined NSCs--all of which have been identified not only by markers but by functional assays in their respective systems and whose state of differentiation could be synchronized--shared a large number of genes. Although, as expected, the most stem-like genes were expressed by ESCs, NSCs and HSCs shared a number of genes. CNS-derived neurospheres, on the other hand, expressed fewer "stem-like" genes held in common by the other operationally-defined stem cell populations. Rather they displayed a profile more consistent with differentiated neural cells. (Genes of neural identity were shared with the NSC clone.) Interestingly, when the operationally-defined NSC clone was cultured as a neurosphere (rather than in monolayer), its expression pattern shifted from a "stem-like" pattern towards a more "differentiated" one, suggesting that the neurosphere, without functional validation, may be a poor model for predicting stem cell attributes because it consists of heterogeneous populations of cells, only a small proportion of which are truly "stem-like". Furthermore, when operational definitions are employed, a common set of stem-like genes does emerge across both embryonic and somatic stem cells of various organ systems, including the nervous system.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Central Nervous System/embryology , Central Nervous System/metabolism , Gene Expression Profiling , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Biomarkers , Cell Line , Cells, Cultured , Central Nervous System/cytology , Clone Cells/cytology , Clone Cells/metabolism , Gene Expression Profiling/statistics & numerical data , Gene Expression Regulation, Developmental/genetics , Humans , Nerve Tissue Proteins/genetics , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Stem Cell Transplantation/methods , Stem Cells/cytology , Stem Cells/metabolism
17.
J Neurosci Res ; 78(4): 455-60, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15468175

ABSTRACT

The publication of a paper entitled "Direct transdifferentiation gives rise to the earliest new hair cells in regenerating avian auditory epithelium" in the Journal of Neuroscience Research offers the opportunity to call attention to a well-developed line of research on the auditory receptor of birds, which should be of interest to students of regeneration and plasticity of the mature nervous system in higher vertebrates, including mammals. Although hair cell proliferation normally stops before hatching, destruction of the auditory receptors of the chicken may be followed by complete regeneration of hair cells. Most of the new hair cells arise from a new wave of proliferation, but Roberson et al. show that about one-third of the new hair cells are formed without undergoing cell division and thus may differentiate from so-called supporting cells or cells with an "intermediate morphology." This finding suggests some models for regeneration of this neuroepithelium, including the possibility that mature supporting cells could transform directly into hair cells. The present Mini-Review discusses some of the models for neural regeneration that future studies might address in the light of our current knowledge and the new report. The possibility is raised that transitional forms of hair cell and supporting cell precursors may reside in the inner ear in a quiescent state until stimulated by damage.


Subject(s)
Ear, Inner/physiology , Models, Neurological , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Animals , Cell Differentiation , Ear, Inner/cytology , Ear, Inner/embryology , Hair Cells, Auditory/embryology , Hair Cells, Auditory/physiology
18.
Curr Opin Otolaryngol Head Neck Surg ; 12(5): 418-25, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15377955

ABSTRACT

PURPOSE OF REVIEW: Sensory hair cells are susceptible to ototoxic damage from a variety of sources, including antibiotic treatment. Unfortunately, this often results in permanent hearing and/or balance problems in humans. By understanding how sensory hair cells die in response to aminoglycoside treatment, preventive strategies may be developed. This review will discuss some of the key recent findings in sensory hair cell death and regeneration. RECENT FINDINGS: Aminoglycosides induce hair cell death through the initiation of apoptosis. Early and late stages of hair cell apoptosis have been defined, and several of the key molecules involved in the cascade have been identified. Moreover, specific inhibitors of apoptosis rescue hair cells from death and preserve function. Hair cell death has been shown to induce regeneration through supporting cell transdifferentiation, proliferation, and new hair cell differentiation in birds and lower vertebrates. Regeneration in the mammalian cochlea does not occur spontaneously, but genetic manipulation of cell cycle genes, induction of new hair cells through gene therapy, and introduction of stem cells into damaged cochleas suggest that repair and replacement of lost hair cells in the organ of Corti may be possible. Finally, continuing investigations of the mouse, zebrafish, and human genomes may one day enable manipulation of the cochlea so that functional regeneration is readily available as a therapeutic intervention. SUMMARY: The discovery that hair cells can regenerate in birds and other nonmammalian vertebrates has fueled a wide range of studies to find ways to restore hearing and balance in mammals. The demonstration that apoptosis and proliferation are coupled as controlling factors in regeneration and the advent of new approaches such as gene therapy, stem cell transplantation, and genomics may lead to methods for inducing hair cell regeneration and repair in the mammalian cochlear and vestibular systems.


Subject(s)
Aminoglycosides/adverse effects , Apoptosis/drug effects , Apoptosis/physiology , Hair Cells, Vestibular/physiology , Regeneration/physiology , Aminoglycosides/therapeutic use , Animals , Ear, Middle/drug effects , Humans , Risk Factors , Sensitivity and Specificity , Signal Transduction , Zebrafish
19.
J Neurosci Res ; 78(4): 461-71, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15372572

ABSTRACT

The avian auditory epithelium is capable of complete regeneration after hair cell (HC) loss. Most new HCs arise via cell division, but approximately one-third of new HCs arise via direct transdifferentiation (DT), in which supporting cells (SCs) alter their phenotype without dividing. In this study, we used synchronous, gentamicin-induced near-total HC loss in the basal end of the epithelium and continuous infusion of the cell division marker bromodeoxyuridine (BrdU) to identify the origin of each individual regenerating HC. Early new HCs were identified by immunolabeling for the HC-specific marker myosin-VIIa, and mitotic cells with BrdU immunolabeling. The first new HCs arising via DT appear 72-96 hr after gentamicin, 24-48 hr earlier than the first new mitotic HCs. After Day 6, however, most new HCs are mitotic. The "intermediate" morphology that has been suggested to be characteristic of DT is seen in HCs arising via both pathways. These findings suggest that DT is a simpler, more rapid process that produces the first new HCs, and that mitotic regeneration is somewhat slower but ultimately produces most new HCs. The identical morphology of regenerating HCs from both pathways suggests that once HC fate is established, all new HCs follow similar cellular processes during differentiation and reorganization into the regenerated epithelium.


Subject(s)
Cell Differentiation/physiology , Epithelium/physiology , Hair Cells, Auditory/physiology , Regeneration/physiology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Division/physiology , Chickens , Dyneins , Gentamicins/toxicity , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Immunohistochemistry/methods , Models, Neurological , Myosin VIIa , Myosins/metabolism , Time Factors
20.
Eur J Neurosci ; 20(8): 2049-58, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15450084

ABSTRACT

Oxidative injury to premyelinating oligodendrocytes (preOLs) in developing white matter has been implicated in the pathogenesis of periventricular leukomalacia, the lesion underlying most cases of cerebral palsy in premature infants. In this study, we investigated the pathways of OL death induced by intracellular glutathione (GSH) depletion. We found that the lipoxygenase (LOX) inhibitors AA-861 and BMD-122 (N-benzyl-N-hydroxy-5-phenylpentamide; BHPP), but not the cyclooxygenase (COX) inhibitor indomethacin, fully protected the cells from GSH depletion caused by cystine deprivation. Arachidonic acid (AA), the substrate for 12-LOX, potentiated the toxicity of mild cystine deprivation and at higher concentration was itself toxic. This toxicity was also blocked by 12-LOX inhibitors. Consistent with a role for 12-LOX in the cell death pathway, 12-LOX activity increased following cystine deprivation in OLs. Blocking 12-LOX with AA-861 effectively inhibited the accumulation of reactive oxygen species (ROS) induced by cystine deprivation. These data suggest that, in OLs, intracellular GSH depletion leads to activation of 12-LOX, ROS accumulation and cell death. Mature OLs were more resistant than preOLs to cystine deprivation. The difference in sensitivity was not due to a difference in 12-LOX activity but rather appeared to be related to the presence of stronger antioxidant defense mechanisms in mature OLs. These results suggest that 12-LOX activation plays a key role in oxidative stress-induced OL death.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonic Acid/pharmacology , Glutathione/metabolism , Oligodendroglia/drug effects , Animals , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Lipoxygenase Inhibitors/pharmacology , Oligodendroglia/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...