Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Phys Med Biol ; 68(20)2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37748504

ABSTRACT

A subcommittee of the Netherlands Commission on Radiation Dosimetry (NCS) was initiated in 2018 with the task to update and extend a previous publication (NCS-15) on the quality assurance of treatment planning systems (TPS) (Bruinviset al2005). The field of treatment planning has changed considerably since 2005. Whereas the focus of the previous report was more on the technical aspects of the TPS, the scope of this report is broader with a focus on a department wide implementation of the TPS. New sections about education, automated planning, information technology (IT) and updates are therefore added. Although the scope is photon therapy, large parts of this report will also apply to all other treatment modalities. This paper is a condensed version of these guidelines; the full version of the report in English is freely available from the NCS website (http://radiationdosimetry.org/ncs/publications). The paper starts with the scope of this report in relation to earlier reports on this subject. Next, general aspects of the commissioning process are addressed, like e.g. project management, education, and safety. It then focusses more on technical aspects such as beam commissioning and patient modeling, dose representation, dose calculation and (automated) plan optimisation. The final chapters deal with IT-related subjects and scripting, and the process of updating or upgrading the TPS.

2.
Soc Psychiatry Psychiatr Epidemiol ; 53(6): 555-566, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29696304

ABSTRACT

PURPOSE: This work complements a quantitative review by Nortje et al. (Lancet Psychiatry 3(2):154-170, 2016) by exploring the qualitative literature in regard to the perceived effectiveness of traditional and faith healing of mental disorders. METHOD: Qualitative studies focusing specifically on traditional and/or faith healing practices for mental illness were retrieved from eight databases. Data were extracted  into basic coding sheets to facilitate the assessment of the quality of eligible papers using the COREQ. RESULTS: Sixteen articles met the inclusion criteria. Despite methodological limitations, there was evidence from the papers that stakeholders perceived traditional and/or faith healing to be effective in treating mental illness, especially when used in combination with biomedical treatment. CONCLUSION: Patients will continue to seek treatment from traditional and/or faith healers for mental illness if they perceive it to be effective regardless of alternative biomedical evidence. This provides opportunities for collaboration to address resource scarcity in low to middle income countries.


Subject(s)
Faith Healing , Medicine, Traditional , Mental Disorders/therapy , Patient Outcome Assessment , Humans , Qualitative Research
3.
Neuroscience ; 316: 261-78, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26718602

ABSTRACT

Cochlear fibrosis is a common finding following cochlear implantation. Evidence suggests that cochlear fibrosis could be triggered by inflammation and epithelial-to-mesenchymal cell transition (EMT). In this study, we investigate the mechanisms of cochlear fibrosis and the risk/benefit ratio of local administration of the anti-inflammatory drug dexamethasone (DEX) and antimitotic drug aracytine (Ara-C). Cochlear fibrosis was evaluated in cochlear fibrosis models of rat cochlear slices in vitro and in KLH-induced immune labyrinthitis and platinum wire cochlear implantation-induced fibrosis in vivo. Cochleae were invaded with tissue containing fibroblastic cells expressing α-SMA (alpha smooth muscle actin), which along with collagen I, fibronectin, and laminin in the extracellular matrix, suggests the involvement of a fibrotic process triggered by EMT in vitro and in vivo. After perilymphatic injection of an adenoviral vector expressing GFP in vivo, we demonstrated that the fibroblastic cells derived from the mesothelial cells of the scalae tympani and vestibuli. Activation of inflammatory and EMT pathways was further assessed by ELISA analysis of the expression of IL-1ß and TGF-ß1. Both markers were elevated in vitro and in vivo, and DEX and Ara-C were able to reduce IL-1ß and TGF-ß1 production. After 5days of culture in vitro, quantification of calcein-positive cells revealed that Ara-C was 30-fold more efficient in preventing fibrosis, and provoked less sensory hair cell loss, than DEX. In KLH-induced immune labyrinthitis and platinum wire-implanted models, Ara-C was more efficient in preventing proliferation of fibrosis with less side effects on hair cells and neurons than DEX. In conclusion, DEX and Ara-C both prevent fibrosis in the cochlea. Analysis of the risk/benefit ratio favors the use of Ara-C for preventing cochlear fibrosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cochlea , Cytokines/metabolism , Wounds and Injuries/complications , Adjuvants, Immunologic/toxicity , Animals , Cochlea/drug effects , Cochlea/injuries , Cochlea/pathology , Cochlea/ultrastructure , Collagen/metabolism , Dexamethasone/pharmacology , Disease Models, Animal , Electrodes, Implanted/adverse effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Fibronectins/metabolism , Fibrosis/drug therapy , Fibrosis/etiology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hemocyanins/toxicity , In Vitro Techniques , Laminin/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Time Factors
4.
Article in English | MEDLINE | ID: mdl-28596897

ABSTRACT

BACKGROUND: Lower and middle income countries (LMICs) are home to >80% of the global population, but mental health researchers and LMIC investigator led publications are concentrated in 10% of LMICs. Increasing research and research outputs, such as in the form of peer reviewed publications, require increased capacity building (CB) opportunities in LMICs. The National Institute of Mental Health (NIMH) initiative, Collaborative Hubs for International Research on Mental Health reaches across five regional 'hubs' established in LMICs, to provide training and support for emerging researchers through hub-specific CB activities. This paper describes the range of CB activities, the process of monitoring, and the early outcomes of CB activities conducted by the five research hubs. METHODS: The indicators used to describe the nature, the monitoring, and the early outcomes of CB activities were developed collectively by the members of an inter-hub CB workgroup representing all five hubs. These indicators included but were not limited to courses, publications, and grants. RESULTS: Results for all indicators demonstrate a wide range of feasible CB activities. The five hubs were successful in providing at least one and the majority several courses; 13 CB recipient-led articles were accepted for publication; and nine grant applications were successful. CONCLUSIONS: The hubs were successful in providing CB recipients with a wide range of CB activities. The challenge remains to ensure ongoing CB of mental health researchers in LMICs, and in particular, to sustain the CB efforts of the five hubs after the termination of NIMH funding.

5.
Br J Pharmacol ; 166(6): 1888-904, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22320124

ABSTRACT

BACKGROUND AND PURPOSE Exposure to an ototoxic level of an aminoglycoside can result in hearing loss. In this we study investigated the otoprotective efficacy of dexamethasone (DXM), melatonin (MLT) and tacrolimus (TCR) in gentamicin (GM)-treated animals and cultures. EXPERIMENTAL APPROACH Wistar rats were divided into controls (treated with saline); exposed to GM only (GM); and three GM-exposed groups treated with either DXM, MLT or TCR. Auditory function and cochlear surface preparations were studied. In vitro studies of oxidative stress, pro-inflammatory cytokine mRNA levels, the MAPK pathway and caspase-3 activation were performed in organ of Corti explants from 3-day-old rats. KEY RESULTS DXM, MLT and TCR decreased levels of reactive oxygen species in GM-exposed explants. The mRNA levels of TNF-α, IL-1ß and TNF-receptor type 1 were significantly reduced in GM + DXM and GM + MLT groups. Phospho-p38 MAPK levels decreased in GM + MLT and GM + TCR groups, while JNK phosphorylation was reduced in GM + DXM and GM + MLT groups. Caspase-3 activation decreased in GM + DXM, GM + MLT and GM + TCR groups. These results were consistent with in vivo results. Local treatment of GM-exposed rat cochleae with either DXM, MLT or TCR preserved auditory function and prevented auditory hair cell loss. CONCLUSIONS AND IMPLICATIONS In organ of Corti explants, GM increased oxidative stress and initiated an inflammatory response that led to the activation of MAPKs and apoptosis of hair cells. The three compounds tested demonstrated otoprotective properties that could be beneficial in the treatment of ototoxicity-induced hearing loss.


Subject(s)
Anti-Bacterial Agents/adverse effects , Dexamethasone/therapeutic use , Gentamicins/adverse effects , Hearing Loss/drug therapy , Melatonin/therapeutic use , Protective Agents/therapeutic use , Tacrolimus/therapeutic use , Animals , Catalase/metabolism , Dexamethasone/pharmacology , Hearing Loss/chemically induced , Hearing Loss/metabolism , Interleukin-1beta/genetics , Male , Melatonin/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Organ of Corti/metabolism , Protective Agents/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Interleukin-1 Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Superoxide Dismutase/metabolism , Tacrolimus/pharmacology , Tumor Necrosis Factor-alpha/genetics
7.
Neuroscience ; 188: 157-67, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21571041

ABSTRACT

The objective was to determine the role of nuclear factor kappa B (NFκB) in dexamethasone base (DXMb) protection of auditory hair cells from tumor necrosis factor-alpha (TNFα)-induced loss on gene expression and cell signaling levels. Organ of Corti (OC) explants from 3-day-old rats were cultured under one of the following conditions: (1) media only--no treatment; (2) media+TNFα; (3) media+TNFα+DXMb; (4) media+TNFα+DXMb+NFκB-Inhibitor (NFκB-I); or (5) media+TNFα+DXMb+NFκBI-Scrambled control (NFκBI-C). A total of 60 organ of Corti explants (OC) were stained with FITC-Phalloidin after 96 h in culture (conditions 1-5) for hair cell counts and imaging of surface characteristics. A total of 108 OC were used for gene expression studies (i.e. B-actin, Bax, Bcl-2, Bcl-xl, and TNFR1) after 0, 24, or 48 h in vitro (conditions 1-4). A total of 86 OC were cultured (conditions 1-3) for 48 h, 36 of which were used for phosphorylated NFκB (p-NFκB) ELISA studies and 50 for whole mount anti-p-NFκB immunostain experiments. TNFα+DXMb exposed cultures demonstrated significant upregulation in anti-apoptotic Bcl-2 and Bcl-xl genes and downregulation in pro-apoptotic Bax gene expression; DXMb treatment of TNFα explants also lowered the Bax/Bcl-2 ratio and inhibited TNFR1 upregulation. After inhibiting NFκB activity with NFκB-I, the gene expression profile following TNFα+DXMb treatment now mimics that of TNFα-challenged OC explants. The levels of p-NFκB and the degree of nuclear translocation are significantly greater in TNFα+DXMb exposed OC explants than observed in the TNFα and control groups in the middle+basal turns of OC explants. These findings were supported by the results of the hair cell counts and the imaging results obtained from the whole mount OC specimens. DXMb protects against TNFα-induced apoptosis of auditory hair cells in vitro via activation of NFκB signaling in hair cell nuclei, and regulation of the expression levels of anti- and pro-apoptotic genes and a pro-inflammatory gene.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Hair Cells, Auditory/drug effects , NF-kappa B/metabolism , Organ of Corti/drug effects , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Organ Culture Techniques , Organ of Corti/metabolism , Organ of Corti/pathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/toxicity
8.
Neuroscience ; 157(2): 405-13, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18838114

ABSTRACT

OBJECTIVE: Determine the molecular mechanism(s) behind tumor necrosis factor-alpha (TNFalpha)-induced loss of auditory hair cells and the ability of dexamethasone base (DXMb) to protect against TNFalpha ototoxicity. METHODS: Hair cell counts: Three-day-old rat organ of Corti explants were cultured under three different conditions: 1) untreated-control; 2) TNFalpha (2 mug/ml); and 3) TNFalpha (2 mug/ml)+DXMb (70 mug/ml) for 4 days, fixed, and stained with FITC-phalloidin. Hair cells were counted in the basal and middle turns. Gene expression: total RNA was extracted from the three different groups of explants at 0, 12, 24 and 48 h. Using quantitative real-time RT-PCR, mRNAs were transcribed into cDNAs and amplification was performed using primers for rat ss-actin (housekeeping gene), TNFR1, Bcl-2, Bax, and Bcl-xl. RESULTS: DXMb protected explant hair cells from TNFalpha-induced loss. Bax gene expression was greater in TNFalpha-exposed explants compared with TNFalpha+DXMb-treated explants at 48 h (P=0.023), confirmed by the increase in the Bax/Bcl-2 ratio at 48 h (P<0.001). These results correlated with increased TNFR1 expression at 24 h (P=0.038). DXMb otoprotection in TNFalpha-exposed cultures was accompanied by an up-regulation of Bcl-xl at both the 24 (P<0.001) and 48 h time points (P=0.030) and up-regulation of Bcl-2 expression at 24 h (P=0.018). DXMb treatment also prevented increases in the expression levels of Bax, TNFR1, and the Bax/Bcl-2 ratio that occurred in untreated TNFalpha-exposed explants. CONCLUSIONS: TNFalpha's ototoxicity may be mediated through an up-regulation of Bax and TNFR1 expression as well as an increase in the Bax/Bcl-2 ratio. DXMb protects the organ of Corti against TNFalpha ototoxicity by up-regulating Bcl-2 and Bcl-xl expression and by inhibiting TNFalpha-induced increases in Bax, TNFR1, and the Bax/Bcl-2 ratio. These results support the use of local dexamethasone treatment to conserve hearing following a trauma.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , Hair Cells, Auditory/drug effects , Organ of Corti/cytology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/genetics , Organ Culture Techniques , Organ of Corti/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Time Factors , Tumor Necrosis Factor-alpha/toxicity , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
9.
Curr Drug Targets CNS Neurol Disord ; 3(4): 325-32, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15379608

ABSTRACT

The c-Jun N-terminal kinases (JNKs) are also called stress activated protein kinases (SAPKs) and are members of the family of mitogen activated protein kinases (MAPKs). While the functions of the JNKs under physiological conditions are diverse and not completely understood, there is increasing evidence that JNKs are potent effectors of apoptosis of oxidative stress-damaged cells in both the brain and the mammalian inner ear following a trauma. The activation of the inducible transcription factor c-Jun by N-terminal phosphorylation is a central event in JNK-mediated apoptosis of oxidative stress-damaged auditory hair cells following exposure to either acoustic trauma or a toxic level of an aminoglycoside antibiotic and also the apoptosis of auditory neurons as a consequence of a loss of the trophic support provided by the auditory hair cells. In this review, we summarise what is known about the expression and activation of G-proteins, JNKs, c-Jun and c-Fos under oxidative stress conditions within the mammalian cochlea. A particular focus is put on a new peptide conjugate that is a promising protective agent(s) and pharmacological strategies for preventing cochlear damage induced by both acoustic trauma and aminoglycoside ototoxic damage.


Subject(s)
Apoptosis/physiology , Deafness/enzymology , MAP Kinase Signaling System/physiology , Nerve Degeneration/enzymology , Oxidative Stress/physiology , Peptides/pharmacology , Aminoglycosides/antagonists & inhibitors , Aminoglycosides/toxicity , Animals , Apoptosis/drug effects , Deafness/drug therapy , Deafness/physiopathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Humans , MAP Kinase Signaling System/drug effects , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Noise/adverse effects , Oxidative Stress/drug effects , Peptides/therapeutic use
10.
Otol Neurotol ; 25(4): 627-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15241246

ABSTRACT

This review covers the general roles of members of the cysteine protease family of caspases in the process of apoptosis (programmed cell death) looking at their participation in both the "extrinsic" cell death receptor and the "intrinsic" mitochondrial cell death pathways. It defines the difference between initiator and effector caspases and shows the progression of caspase activations that ends up in the apoptotic cell death and elimination of a damaged cell. The review then presents what is currently know about the participation of caspases in the programmed cell death of inner ear sensory cells during the process of normal development and maturation of the inner ear and their importance in this process as illustrated by the results of caspase-3 gene knockout experiments. The participation of specific caspases and the sequence of their activation in the elimination (apoptosis) of damaged sensory cells from adult inner ears after an injury that generates oxidative stress are reviewed. Both the possibility and the potential efficacy of caspase inhibition with a broad-spectrum pancaspase inhibitor as an interventional therapy to treat and rescue oxidative stress-damaged inner ear sensory cells from apoptosis are presented and discussed.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Hair Cells, Auditory/pathology , Oxidative Stress/physiology , Adult , Aging/pathology , Aging/physiology , Animals , Caspase Inhibitors , Caspases/genetics , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Gerbillinae , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/pathology , Humans , Mice , Mice, Knockout , Mitochondria/physiology
11.
J Neurosci ; 23(24): 8596-607, 2003 Sep 17.
Article in English | MEDLINE | ID: mdl-13679429

ABSTRACT

Hearing loss can be caused by a variety of insults, including acoustic trauma and exposure to ototoxins, that principally effect the viability of sensory hair cells via the MAP kinase (MAPK) cell death signaling pathway that incorporates c-Jun N-terminal kinase (JNK). We evaluated the otoprotective efficacy of D-JNKI-1, a cell permeable peptide that blocks the MAPK-JNK signal pathway. The experimental studies included organ cultures of neonatal mouse cochlea exposed to an ototoxic drug and cochleae of adult guinea pigs that were exposed to either an ototoxic drug or acoustic trauma. Results obtained from the organ of Corti explants demonstrated that the MAPK-JNK signal pathway is associated with injury and that blocking of this signal pathway prevented apoptosis in areas of aminoglycoside damage. Treatment of the neomycin-exposed organ of Corti explants with D-JNKI-1 completely prevented hair cell death initiated by this ototoxin. Results from in vivo studies showed that direct application of D-JNKI-1 into the scala tympani of the guinea pig cochlea prevented nearly all hair cell death and permanent hearing loss induced by neomycin ototoxicity. Local delivery of D-JNKI-1 also prevented acoustic trauma-induced permanent hearing loss in a dose-dependent manner. These results indicate that the MAPK-JNK signal pathway is involved in both ototoxicity and acoustic trauma-induced hair cell loss and permanent hearing loss. Blocking this signal pathway with D-JNKI-1 is of potential therapeutic value for long-term protection of both the morphological integrity and physiological function of the organ of Corti during times of oxidative stress.


Subject(s)
Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/drug effects , Hearing Loss, Noise-Induced/prevention & control , Hearing Loss/prevention & control , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Organ of Corti/drug effects , Peptides/pharmacology , Acoustic Stimulation , Aminoglycosides/antagonists & inhibitors , Aminoglycosides/toxicity , Animals , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Evaluation, Preclinical , Guinea Pigs , Hair Cells, Auditory/cytology , Hearing Loss/chemically induced , Hearing Tests , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases , Ligands , Mice , Neuroprotective Agents/pharmacology , Organ of Corti/cytology , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects
12.
J Neurochem ; 87(2): 508-21, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14511128

ABSTRACT

In the current study, we have investigated the ability of substance P (SP) to protect 3-day-old (P3) rat spiral ganglion neurons (SGNs) from trophic factor deprivation (TFD)-induced cell death. The presence of SP high affinity neurokinin-1 receptor (NK1) transcripts was detected in the spiral ganglion and the NK1 protein localized to SGNs both ex vivo and in vitro. Treatment with SP increased cytoplasmic Ca2+ in SGNs, further arguing for the presence of functional NK1 on these neurons. Both SP and the agonist [Sar9,Met(O2)11]-SP significantly decreased SGN cell death induced by TFD, with no effect on neurite outgrowth. The survival promoting effect of SP was blocked by the NK1 antagonist, WIN51708. Both pan-caspase inhibitor BOC-D-FMK and SP treatments markedly reduced activation of caspases and DNA fragmentation in trophic factor deprived-neurons. The neuroprotective action of SP was antagonised by specific inhibitors of second messengers, including 1.2-bis-(O-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM) to chelate cytosolic Ca2+, the protein kinase C (PKC) inhibitors bisindolylmaleimide I, Gö6976 and LY333531 and the MAPK/ERK inhibitor U0126. In contrast, nifedipine, a specific inhibitor of l-type Ca2+ channel, and LY294002, a phosphatidylinositol-3-OH kinase (PI3K) inhibitor, had no effect on SP trophic support of SGNs. Moreover, activation of endogenous PKC by 4 beta-phorbol 12-myristate 13-acetate (PMA) also reduced the loss of trophic factor-deprived SGNs. Thus, NK1 expressed by SGNs transmit a survival-promoting regulatory signal during TFD-induced SGN cell death via pathways involving PKC activation, Ca2+ signalling and MAPK/ERK activation, which can be accounted for by an inhibition of caspase activation.


Subject(s)
Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Spiral Ganglion , Substance P/pharmacology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Cytosol/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/pharmacology , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nifedipine/pharmacology , Protein Kinase C/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Substance P/biosynthesis , Substance P/genetics
13.
Cell Mol Life Sci ; 59(10): 1744-57, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12475185

ABSTRACT

The organ of Corti is a complex structure containing a single row of inner hair cells (IHCs) and three rows of outer hair cells (OHCs), supported respectively by one row of inner phalangeal cells and three rows of Deiters' cells. When fetal rat organ of Corti explants are cultured, supernumerary OHCs and supernumerary Deiters' cells are produced, without any additional cell proliferation. Analysis of semi- and ultrathin sections revealed that supernumerary OHCs are produced at the distal edge of the organ of Corti. Quantitative analysis of cell types present in the organ of Corti demonstrates that when the number of OHCs increases: (i) the total number of cells remains constant; (ii) the number of Deiters' cells increases; (iii) the number of tectal cells decreases and of Hensen's cells decreases. Using specific HC markers, i.e. jagged2 (Jag2) and Math1, we showed that in addition to existing OHCs, supernumerary OHCs, tectal cells and Hensen's cells expressed these markers in embryonic day 19 organ of Corti explants after 5 days in vitro. The results of this study suggest that Hensen's cells retain the capacity to differentiate into either tectal cells, which differentiate into OHCs, or into undertectal cells which differentiate into Deiters' cells.


Subject(s)
Cell Differentiation/physiology , Embryonic and Fetal Development/physiology , Epithelial Cells/cytology , Hair Cells, Auditory, Outer/embryology , Organ of Corti/embryology , Vestibular Nucleus, Lateral/embryology , Animals , Epithelial Cells/physiology , Female , Hair Cells, Auditory, Outer/cytology , Organ Culture Techniques , Organ of Corti/cytology , Pregnancy , Rats , Rats, Wistar , Vestibular Nucleus, Lateral/cytology
14.
Acta Otorhinolaryngol Belg ; 56(1): 45-9, 2002.
Article in English | MEDLINE | ID: mdl-11894630

ABSTRACT

Most of the deafness are of sensorineural origin and are characterized by a loss of hair cells and of spiral ganglion neurons. At the present time, hearing aids are the only treatment. However, in some diseases of the inner ear, pharmacological treatment have been proposed and used successfully. In this paper, we will review some basic science aspects of the biology of the neurosensory structures of the inner ear, in particular of the auditory neurons, that lead to the rationale of some treatments for the inner ear diseases. Developmental studies, neuronal cell culture experiments, and analyses of gene knockout animals reveal a number of growth factors which are important for the rescue and repair of injured auditory neurons in the inner ear. These factors rescue the injured auditory neurons in vivo. Furthermore, perfusion of antioxydant to the cochlea prevented the hearing loss induced by cisplatin. These in vitro and in vivo experiments demonstrate that it is possible to manipulate the neurosensory structures of the inner ear and provide an effective treatment to prevent the degeneration of the neurons. The molecules or drugs can be administered locally to the inner ear through a direct perilymphatic perfusion or through the round window membrane. As an example, we will discuss the treatment of patients suffering from idiopathic sensorineural hearing loss which can be treated successfully by a perfusion through the round window membrane, improving their hearing threshold and their speech discrimination.


Subject(s)
Ear, Inner/drug effects , Ear, Inner/innervation , Labyrinth Diseases/drug therapy , Animals , Cochlear Nerve/drug effects , Cochlear Nerve/physiopathology , Disease Models, Animal , Ear, Inner/physiopathology , Humans , In Vitro Techniques , Labyrinth Diseases/physiopathology , Mice , Rats
15.
Development ; 128(24): 5017-29, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748138

ABSTRACT

Development of the vertebrate inner ear is characterized by a series of genetically programmed events involving induction of surface ectoderm, preliminary morphogenesis, specification and commitment of sensory, nonsensory and neuronal cells, as well as outgrowth and restructuring of the otocyst to form a complex labyrinth. Hmx2, a member of the Hmx homeobox gene family, is coexpressed with Hmx3 in the dorsolateral otic epithelium. Targeted disruption of Hmx2 in mice demonstrates the temporal and spatial involvement of Hmx2 in the embryonic transition of the dorsal portion (pars superior) of the otocyst to a fully developed vestibular system. In Hmx2 null embryos, a perturbation in cell fate determination in the lateral aspect of the otic epithelium results in reduced cell proliferation in epithelial cells, which includes the vestibular sensory patches and semicircular duct fusion plates, as well as in the adjacent mesenchyme. Consequently, enlargement and morphogenesis of the pars superior of the otocyst to form a complex labyrinth of cavities and ducts is blocked, as indicated by the lack of any distinguishable semicircular ducts, persistence of the primordial vestibular diverticula, significant loss in the three cristae and the macula utriculus, and a fused utriculosaccular chamber. The developmental regulators Bmp4, Dlx5 and Pax2 all play a critical role in inner ear ontogeny, and the expression of each of these genes is affected in the Hmx2 null otocyst suggesting a complex regulatory role for Hmx2 in this genetic cascade. Both Hmx2 and Hmx3 transcripts are coexpressed in the developing central nervous system including the neural tube and hypothalamus. A lack of defects in the CNS, coupled with the fact that not all of the Hmx2-positive regions in developing inner ear are impaired in the Hmx2 null mice, suggest that Hmx2 and Hmx3 have both unique and overlapping functions during embryogenesis.


Subject(s)
Drosophila Proteins , Genes, Homeobox , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Transcription Factors , Vestibule, Labyrinth/embryology , Animals , Behavior, Animal , Cell Division , Epithelial Cells/cytology , Gene Expression Regulation, Developmental , Genes, Reporter , Hair Cells, Auditory/cytology , Heterozygote , Homozygote , Hyperkinesis/genetics , Mesoderm/cytology , Mice , Mice, Mutant Strains , Morphogenesis , Mutagenesis, Insertional , Saccule and Utricle/cytology , Vestibule, Labyrinth/abnormalities , Vestibule, Labyrinth/innervation
16.
Acta Otolaryngol ; 121(6): 666-72, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11678164

ABSTRACT

Presbycusis is a complex of high frequency hearing loss and disproportionate loss of speech discrimination that is seen concomitantly with physical signs of aging. Among the most extensively characterized strains of mice that show an early hearing loss is the C57B16/J strain, a strain that shows early onset of high frequency hearing loss at age 6 months and complete hearing loss by 1 year of age. The histopathology of this strain consists of loss of hair cells and spiral ganglion neurons in the basal turn, with a progression of loss of hair cells and ganglion neurons towards the apical portion of the cochlea as the animal ages. The process of aging has been extensively studied and although details differ in various organisms the consensus today is that oxidative stress, i.e. free radical-mediated tissue damage, is one of the core mechanisms of aging. Aerobic metabolism results in the creation of hydrogen peroxide and reactive oxygen species. These are normally detoxified by a variety of enzymes and free radical scavengers, including superoxide dismutase (SOD), catalase and glutathione. To determine whether oxidative stress plays a role in the pathophysiology of hearing loss in this mouse model of presbycusis we determined the relative change in mRNA production for selected free radical detoxifying enzymes in the C57B16/J mouse cochlea. Using semi-quantitative RT-PCR with tubulin mRNA as a control, relative levels of antioxidant enzyme mRNAs were determined. There was an overall increase in SOD1 mRNA levels when comparing 1 and 9 month time points, and a transient increase in the expression level of catalase mRNA. B6.CAST+ Ahl mice, which carry the C57B16/J genome but receive their Ahl gene from CAST mice, do not show these alteractions in antioxidant enzyme production. Our results suggest that at an age of 9 months, at which point significant hearing loss has developed, the C57B16/J mouse cochlea is exposed to increased levels of free radicals and that the Ahl gene of the C57B16/J mouse mediates this decrease in protective enzymes and therefore increase in levels of oxidative stress.


Subject(s)
Aging/physiology , Cochlea/metabolism , Oxidative Stress/physiology , Animals , Catalase/metabolism , Cochlea/cytology , Cochlea/physiopathology , Glutathione Peroxidase/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Hearing Disorders/physiopathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
17.
Laryngoscope ; 111(7): 1147-55, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11568534

ABSTRACT

OBJECTIVES: The aim of this study is to determine the efficacy of L-N-acetyl-cysteine (L-NAC) as a protectant for inner ear auditory sensory cells against the toxic effects of cisplatin. STUDY DESIGN: Prospective laboratory study of the otoprotective effect of L-NAC on auditory neurons and hair cells in vitro. METHODS: The study has two arms. The first arm evaluated the neuroprotective effect of L-NAC on early postpartum auditory ganglion cell cultures. Two culture media were used. The two media differed in that one of them was enhanced by the addition of neurotrophins (neurotrophin type 3 and brain-derived neurotrophic factor) and a growth factor (transforming growth factor-beta1). Then the survival of cisplatin-treated auditory neurons was studied before and after pretreatment with protective levels of L-NAC. The second arm of the study evaluated the effect of L-NAC on cisplatin damage initiated to auditory hair cells. Early-postpartum organ of Corti explants were grown in culture. Their rate of survival was studied after exposure to toxic levels of cisplatin. Then, survival of cisplatin-damaged hair cells was studied after they were pretreated with L-NAC. RESULTS: Pretreatment of cultures with L-NAC protected both auditory neurons and hair cells from the effects of exposure to toxic levels of cisplatin. This observed otoprotective effect was dose dependent. CONCLUSIONS: Our in vitro studies have demonstrated that L-NAC protected both auditory neurons and hair cells from the toxic effects of cisplatin. Because it protects both of these inner ear structures, L-NAC may be potentially useful in protecting hearing, in general, from cisplatin-induced damage. In addition, L-NAC has low systemic and mucosal toxicity. It also has a low molecular weight that may allow it to readily cross the round window membrane. All these characteristics make it potentially suitable for transtympanic application for the prevention of the ototoxicity of cisplatin in vivo.


Subject(s)
Acetylcysteine/pharmacology , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlear Nerve/drug effects , Free Radical Scavengers/pharmacology , Hair Cells, Auditory, Inner/drug effects , Neurons/drug effects , Analysis of Variance , Animals , Cells, Cultured , Deafness/chemically induced , Deafness/prevention & control , Hearing/drug effects , Humans , Organ of Corti/drug effects , Prospective Studies , Rats , Time Factors
19.
Neurotoxicology ; 22(2): 163-76, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11405249

ABSTRACT

Cisplatin (cis-diamminedichloroplatinum(II) (CDDP)) is a widely used, highly effective, oncolytic agent that has serious ototoxic side-effects. To test the effectiveness of local delivery, of L-methionine (L-Met) as an otoprotective agent against CDDP ototoxicity, we used a rat model of a highly metastatic breast cancer tumor, i.e. Fisher 344 rats implanted with MTLn3 breast cancer cells. Four experimental groups were evaluated--I: untreated; II: CDDP-treated (three dosages); III: systemically-delivered L-Met + CDDP-treated; IV: locally delivered L-Met + CDDP-treated. The integrity of the outer hair cells (OHCs) was determined using scanning electron microscopy (SEM); hearing was assessed by recording auditory brainstem responses (ABRs) at multiple frequencies. The chemotherapeutic effectiveness of CDDP was quantified by measuring changes in tumor mass and the presence of tumor metastasis. L-Met provided otoprotection of the OHCs against CDDP toxicity in the cochleae of rats following either systemic (III) or local (IV) administration. The ABRs were unchanged in each of the L-Met protection Groups (III and IV) and in the untreated animals of Group I. Treatment with CDDP only (II) induced significant hearing losses at both 16 and 18 kHz when compared to ABRs of untreated rats(I). CDDP was effective in controlling the MTLn3 initiated breast cancer tumors in the CDDP-treated (II) and the local L-Met protection, CDDP-treated (IV) Groups. In contrast, the tumors in the systemic L-Met protection, CDDP-treated Group (III) were not controlled by the CDDP treatment regime. This study demonstrates that local delivery of L-Met to the scala tympani of the cochlea via the round window membrane (IV) provides effective protection against CDDP ototoxicity without compromising its ability to control a highly metastatic form of cancer.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/antagonists & inhibitors , Cisplatin/toxicity , Hearing Disorders/chemically induced , Hearing Disorders/prevention & control , Methionine/administration & dosage , Methionine/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Implants , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Hair Cells, Auditory, Outer/pathology , Hearing Disorders/pathology , Injections, Intraperitoneal , Membranes, Artificial , Microscopy, Electron, Scanning , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
20.
Acta Otolaryngol ; 121(2): 157-63, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11349769

ABSTRACT

Successful delivery of genes to the inner ear has been demonstrated using a variety of vectors and animal models. As our understanding of the molecular pathophysiology of hearing and balance disorders increases, the delivery of genes is becoming central to our ability to manipulate the function of the inner ear. This study evaluates the efficacy of gene transfer and the distribution of three different vector types within the inner ear. Adenovirus vectors, herpes virus vectors and liposomes carrying a plasmid with the green fluorescent protein or beta galactosidase marker genes and a CMV promoter were introduced into the inner ear of 3-month-old mice. The temporal bones and brain were then removed from the animals and examined for transgene expression. Distribution of staining in the treated ear was compared with distribution of staining in the contralateral inner ear. Staining for T cell markers was also carried out to determine inner ear immune response to gene transfer. Herpes virus vectors appear to target neurons most efficiently. Liposome vectors were least efficient in terms of gene transfer. Adenovirus vectors accomplished gene transfer to the widest variety of inner ear cells including auditory and vestibular hair cells. Newer generation adenovirus vectors promise less immune reaction and toxicity than traditional vectors and will be useful for both research and future clinical applications.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Hearing Loss, Sensorineural/therapy , Animals , Cochlea/pathology , Female , Gene Expression/physiology , Genetic Markers/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Mice , Neurons/pathology , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...