Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Neurol ; 14: 1268359, 2023.
Article in English | MEDLINE | ID: mdl-37885485

ABSTRACT

Introduction: Vestibular schwannoma (VS) is an intracranial tumor that arises on the vestibular branch of cranial nerve VIII and typically presents with sensorineural hearing loss (SNHL). The mechanisms of this SNHL are postulated to involve alterations in the inner ear's microenvironment mediated by the genetic cargo of VS-secreted extracellular vesicles (EVs). We aimed to identify the EV cargo associated with poor hearing and determine whether its delivery caused hearing loss and cochlear damage in a mouse model in vivo. Methods: VS tissue was collected from routinely resected tumors of patients with good (VS-GH) or poor (VS-PH) pre-surgical hearing measured via pure-tone average and word recognition scores. Next-generation sequencing was performed on RNA isolated from cultured primary human VS cells and EVs from VS-conditioned media, stratified by patients' hearing ability. microRNA expression levels were compared between VS-PH and VS-GH samples to identify differentially expressed candidates for packaging into a synthetic adeno-associated viral vector (Anc80L65). Viral vectors containing candidate microRNA were infused to the semicircular canals of mice to evaluate the effects on hearing, including after noise exposure. Results: Differentially expressed microRNAs included hsa-miR-431-5p (enriched in VS-PH) and hsa-miR-192-5p (enriched in VS-GH). Newborn mice receiving intracochlear injection of viral vectors over-expressing hsa-miR-431-GFP, hsa-miR-192-GFP, or GFP only (control) had similar hearing 6 weeks post-injection. However, after acoustic trauma, the miR-431 group displayed significantly worse hearing, and greater loss of synaptic ribbons per inner hair cell in the acoustically traumatized cochlear region than the control group. Conclusion: Our results suggest that miR-431 contributes to VS-associated hearing loss following cochlear stress. Further investigation is needed to determine whether miR-431 is a potential therapeutic target for SNHL.

2.
Bone ; 145: 115837, 2021 04.
Article in English | MEDLINE | ID: mdl-33385614

ABSTRACT

The bone encasing the inner ear, known as the otic capsule, is unique because it remodels little postnatally compared to other bones in the body. Previous studies established that osteoprotegerin (OPG) in the inner ear inhibits otic capsule remodeling. OPG acts as a decoy receptor of receptor activator of nuclear factor κB ligand (RANKL) to disrupt the interaction between RANKL and RANK, the primary regulators of bone metabolism. Here we studied the expression and function of RANK and RANKL in the murine cochlea. Using a combination of in situ hybridization, real-time quantitative RT-PCR, and western blot, we demonstrate that Rankl and Rank genes and their protein products are expressed in the intracochlear soft tissues and the otic capsule in a developmentally regulated manner. Using a culture of neonatal murine cochlear neurons, we show that the interaction between RANK and RANKL inhibits neurite outgrowth in these neurons, and is associated with upregulation of NOGO-A expression. Taken together, our results suggest that, in addition to regulating otic capsule bone remodeling, RANK and RANKL expressed by intracochlear soft tissues may also regulate spiral ganglion neuron function by affecting neurite outgrowth.


Subject(s)
Ear, Inner , RANK Ligand , Animals , Bone Remodeling , Mice , Nogo Proteins , Osteoprotegerin/genetics , Receptor Activator of Nuclear Factor-kappa B
3.
PLoS One ; 14(6): e0218292, 2019.
Article in English | MEDLINE | ID: mdl-31185063

ABSTRACT

The vast majority of hearing loss, the most common sensory impairment, and vertigo, which commonly causes falls, both reflect underlying dysfunction of inner ear cells. Perilymph sampling can thus provide molecular cues to hearing and balance disorders. While such "liquid biopsy" of the inner ear is not yet in routine clinical practice, previous studies have uncovered alterations in perilymph in patients with certain types of hearing loss. However, the proteome of perilymph from patients with intact hearing has been unknown. Furthermore, no complete characterization of perilymph from patients with vestibular dysfunction has been reported. Here, using liquid-chromatography with tandem mass spectrometry, we analyzed samples of normal perilymph collected from three patients with skull base meningiomas and intact hearing. We identified 228 proteins that were common across the samples, establishing a greatly expanded proteome of the previously inferred normal human perilymph. Further comparison to perilymph obtained from three patients with vestibular dysfunction with drop attacks due to Meniere's disease showed 38 proteins with significantly differential abundance. The abundance of four protein candidates with previously unknown roles in inner ear biology was validated in murine cochleae by immunohistochemistry and in situ hybridization: AACT, HGFAC, EFEMP1, and TGFBI. Together, these results motivate future work in characterizing the normal human perilymph and identifying biomarkers of inner ear disease.


Subject(s)
Cochlea/metabolism , Meniere Disease/metabolism , Perilymph/metabolism , Proteome/metabolism , Vertigo/metabolism , Animals , Biomarkers/metabolism , Chromatography, Liquid , Cochlea/pathology , Female , Humans , Male , Meniere Disease/pathology , Mice , Middle Aged , Tandem Mass Spectrometry , Vertigo/pathology
4.
Dev Neurobiol ; 76(12): 1374-1386, 2016 12.
Article in English | MEDLINE | ID: mdl-27112738

ABSTRACT

Noise-induced hearing loss (NIHL) is a problem of profound clinical significance and growing magnitude. Alarmingly, even moderate noise levels, previously assumed to cause only temporary shifts in auditory thresholds ("temporary" NIHL), are now known to cause cochlear synaptopathy and subsequent neuropathy. To uncover molecular mechanisms of this neuropathy, a network analysis of genes reported to have significantly altered expression after temporary threshold shift-inducing noise exposure was performed. The transcription factor Hepatocyte Nuclear Factor-4 alpha (HNF4α), which had not previously been studied in the context of cochlear response to noise, was identified as a hub of a top-ranking network. Hnf4α expression and localization using quantitative RT-PCR and in situ hybridization, respectively, were described in adolescent and adult mice exposed to neuropathic noise levels in adolescence. Isoforms α3 and α12 in the cochlea were also identified. At every age examined, Hnf4α mRNA expression in the cochlear apex was similar to expression in the base. Hnf4α expression was evident in select cochlear cells, including spiral ganglion neurons (SGNs) and hair cells, and was significantly upregulated from 6 to 70 weeks of age, especially in SGNs. This age-related Hnf4α upregulation was inhibited by neuropathic noise exposure in adolescence. Hnf4α silencing with shRNA transfection into auditory neuroblast cells (VOT-33) reduced cell viability, as measured with the MTT assay, suggesting that Hnf4α may be involved in SGN survival. Our results motivate future studies of HNF4α in cochlear pathophysiology, especially because HNF4α mutations and polymorphisms are associated with human diseases that may include hearing loss. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1374-1386, 2016.


Subject(s)
Auditory Threshold/physiology , Cochlea/physiology , Hearing Loss, Noise-Induced/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Noise , Animals , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/physiopathology , Hepatocyte Nuclear Factor 4/genetics , Mice
5.
Aging Cell ; 15(2): 301-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26791792

ABSTRACT

Tumor necrosis factor (TNF) family cytokines are important mediators of inflammation. Elevated levels of serum TNF-α are associated with human sensorineural hearing loss via poorly understood mechanisms. We demonstrate, for the first time, expression of TNF-related apoptosis-inducing ligand (TRAIL) and its signaling death receptor 5 (DR5) in the murine inner ear and show that exogenous TRAIL can trigger hair cell and neuronal degeneration, which can be partly prevented with DR5-blocking antibodies.


Subject(s)
Hair Cells, Auditory, Inner/metabolism , Hearing Loss, Sensorineural/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cochlea/metabolism , Cochlea/pathology , Ear, Inner/metabolism , Ear, Inner/pathology , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/therapy , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology
6.
Sci Rep ; 5: 12479, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26216189

ABSTRACT

Osteoprotegerin (OPG) is a key regulator of bone remodeling. Mutations in OPG are involved in a variety of human diseases. We have shown that cochlear spiral ganglion cells secrete OPG at high levels and lack of OPG causes sensorineural hearing loss in addition to the previously described conductive hearing loss. In order to study the regulation of OPG expression, we conducted a database search on regulatory elements in the promoter region of the OPG gene, and identified two potential GATA-3 binding sites. Using luciferase assays and site directed mutagenesis, we demonstrate that these two elements are GATA-3 responsive and support GATA-3 transactivation in human HEK and HeLa cells. The expression of wild type GATA-3 activated OPG mRNA and protein expression, while the expression of a dominant negative mutant of GATA-3 or a GATA-3 shRNA construct reduced OPG mRNA and protein levels. GATA-3 deficient cells generated by expressing a GATA-3 shRNA construct were sensitive to apoptosis induced by etoposide and TNF-α. This apoptotic effect could be partly prevented by the co-treatment with exogenous OPG. Our results suggest new approaches to rescue diseases due to GATA-3 deficiency - such as in hypoparathyroidism, sensorineural deafness, and renal (HDR) syndrome - by OPG therapy.


Subject(s)
Osteoprotegerin/genetics , Promoter Regions, Genetic , Transcriptional Activation , GATA3 Transcription Factor/physiology , HEK293 Cells , Humans
7.
Mol Oncol ; 9(7): 1359-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25891780

ABSTRACT

Vestibular schwannomas (VSs), the most common tumors of the cerebellopontine angle, arise from Schwann cells lining the vestibular nerve. Pharmacotherapies against VS are almost non-existent. Although the therapeutic inhibition of inflammatory modulators has been established for other neoplasms, it has not been explored in VS. A bioinformatic network analysis of all genes reported to be differentially expressed in human VS revealed a pro-inflammatory transcription factor nuclear factor-kappa B (NF-κB) as a central molecule in VS pathobiology. Assessed at the transcriptional and translational level, canonical NF-κB complex was aberrantly activated in human VS and derived VS cultures in comparison to control nerves and Schwann cells, respectively. Cultured primary VS cells and VS-derived human cell line HEI-193 were treated with specific NF-κB siRNAs, experimental NF-κB inhibitor BAY11-7082 (BAY11) and clinically relevant NF-κB inhibitor curcumin. Healthy human control Schwann cells from the great auricular nerve were also treated with BAY11 and curcumin to assess toxicity. All three treatments significantly reduced proliferation in primary VS cultures and HEI-193 cells, with siRNA, 5 µM BAY11 and 50 µM curcumin reducing average proliferation (±standard error of mean) to 62.33% ± 10.59%, 14.3 ± 9.7%, and 23.0 ± 20.9% of control primary VS cells, respectively. These treatments also induced substantial cell death. Curcumin, unlike BAY11, also affected primary Schwann cells. This work highlights NF-κB as a key modulator in VS cell proliferation and survival and demonstrates therapeutic efficacy of directly targeting NF-κB in VS.


Subject(s)
NF-kappa B/antagonists & inhibitors , Neurilemmoma/therapy , Vestibular Diseases/therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Curcumin/pharmacology , Gene Knockdown Techniques , Humans , NF-kappa B/genetics , Neurilemmoma/metabolism , Neurilemmoma/pathology , Vestibular Diseases/metabolism , Vestibular Diseases/pathology
8.
Transl Res ; 166(1): 1-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25616959

ABSTRACT

Vestibular schwannomas (VSs) are the most common tumors of the cerebellopontine angle. Significant clinical need exists for pharmacotherapies against VSs. Motivated by previous findings that immunohistochemical expression of cyclooxygenase 2 (COX-2) correlates with VS growth rate, we investigated the role of COX-2 in VSs and tested COX-2 inhibiting salicylates against VSs. COX-2 was found to be aberrantly expressed in human VS and primary human VS cells in comparison with control human nerve specimens and primary Schwann cells (SCs), respectively. Furthermore, levels of prostaglandin E2, the downstream enzymatic product of COX-2, were correlated with primary VS culture proliferation rate. Because COX-2 inhibiting salicylates such as aspirin are well tolerated and frequently clinically used, we assessed their repurposing for VS. Changes in proliferation, cell death, and cell viability were analyzed in primary VS cultures treated with aspirin, sodium salicylate, or 5-aminosalicylic acid. These drugs neither increased VS cell death nor affected healthy SCs. The cytostatic effect of aspirin in vitro was in concurrence with our previous clinical finding that patients with VS taking aspirin demonstrate reduced tumor growth. Overall, this work suggests that COX-2 is a key modulator in VS cell proliferation and survival and highlights salicylates as promising pharmacotherapies against VS.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cytostatic Agents/pharmacology , Neuroma, Acoustic/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Gene Expression/drug effects , Humans , Immunohistochemistry , Mesalamine/pharmacology , Neuroma, Acoustic/metabolism , Neuroma, Acoustic/pathology , Sodium Salicylate/pharmacology , Translational Research, Biomedical , Tumor Cells, Cultured
9.
Neurobiol Dis ; 56: 25-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23607938

ABSTRACT

Osteoprotegerin (OPG) is a key regulator of bone remodeling. Mutations and variations in the OPG gene cause many human diseases that are characterized by not only skeletal abnormalities but also poorly understood hearing loss: Paget's disease, osteoporosis, and celiac disease. To gain insight into the mechanisms of hearing loss in OPG deficiency, we studied OPG knockout (Opg(-/-)) mice. We show that they develop sensorineural hearing loss, in addition to conductive hearing loss due to abnormal middle-ear bones. OPG deficiency caused demyelination and degeneration of the cochlear nerve in vivo. It also activated ERK, sensitized spiral ganglion cells (SGC) to apoptosis, and inhibited proliferation and survival of cochlear stem cells in vitro, which could be rescued by treatment with exogenous OPG, an ERK inhibitor, or bisphosphonate. Our results demonstrate a novel role for OPG in the regulation of SGC survival, and suggest a mechanism for sensorineural hearing loss in OPG deficiency.


Subject(s)
Cochlear Nerve/pathology , Ear, Inner/pathology , Hearing Loss, Sensorineural/pathology , Nerve Degeneration/pathology , Osteoprotegerin/biosynthesis , Vestibulocochlear Nerve Diseases/pathology , Animals , Apoptosis/physiology , Cell Survival , Cells, Cultured , Cochlear Nerve/metabolism , Ear, Inner/metabolism , Enzyme-Linked Immunosorbent Assay , Hearing Loss, Sensorineural/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Osteoprotegerin/genetics , Oxidative Stress/physiology , Paraffin Embedding , Phenotype , Plastic Embedding , Schwann Cells/metabolism , Spiral Ganglion/cytology , Vestibulocochlear Nerve Diseases/metabolism
10.
J Proteome Res ; 10(9): 3845-51, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21740021

ABSTRACT

Current diagnostic tools limit a clinician's ability to discriminate between many possible causes of sensorineural hearing loss. This constraint leads to the frequent diagnosis of the idiopathic condition, leaving patients without a clear prognosis and only general treatment options. As a first step toward developing new diagnostic tools and improving patient care, we report the first use of liquid chromatography-tandem mass-spectrometry (LC-MS/MS) to map the proteome of human perilymph. Using LC-MS/MS, we analyzed four samples, two collected from patients with vestibular schwannoma (VS) and two from patients undergoing cochlear implantation (CI). For each cohort, one sample contained pooled specimens collected from five patients and the second contained a specimen obtained from a single patient. Of the 271 proteins identified with high confidence among the samples, 71 proteins were common in every sample and used to conservatively define the proteome of human perilymph. Comparison to human cerebrospinal fluid and blood plasma, as well as murine perilymph, showed significant similarity in protein content across fluids; however, a quantitative comparison was not possible. Fifteen candidate biomarkers of VS were identified by comparing VS and CI samples. This list will be used in future investigations targeted at discriminating between VS tumors associated with good versus poor hearing.


Subject(s)
Perilymph/chemistry , Proteome/analysis , Biomarkers/analysis , Biomarkers/chemistry , Cerebrospinal Fluid/chemistry , Chromatography, Liquid , Cochlear Implantation , Cohort Studies , Humans , Neuroma, Acoustic/metabolism , Peptide Fragments/analysis , Peptide Fragments/chemistry , Plasma/chemistry , Proteome/chemistry , Proteomics/methods , Tandem Mass Spectrometry
11.
Otolaryngol Head Neck Surg ; 145(1): 171-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21493287

ABSTRACT

OBJECTIVE: To determine whether variations in gene expression exist at multiple subsites along the sinonasal tract in patients with chronic sinusitis with polyps and in healthy controls. STUDY DESIGN: Prospective, controlled study. SETTING: Academic medical center. SUBJECTS AND METHODS: Tissue expression levels of 5 genes, previously found to be characteristic of ethmoid polyps, were measured using real-time quantitative polymerase chain reaction in 100 sinonasal tissue samples. Specimens harvested from 5 regions--the ethmoid sinus, septum, inferior turbinate, middle turbinate, and lateral nasal wall--in 10 patients with chronic sinusitis and ethmoid polyps were compared to tissue from similar regions in 10 control patients without sinusitis. Western blot analysis was performed to validate differential gene expression at the protein level. RESULTS: Gene expression levels of ethmoid polyps differed significantly from those of healthy ethmoid mucosa, as well as tissue from 4 surrounding anatomical sites in both patients with chronic sinusitis and controls. Alterations specific to the polyp tissue included downregulated genes, prolactin-induced protein (fold change 377.2 ± 169.0, P < .0001), and zinc α2-glycoprotein (fold change 72.1 ± 26.5, P < .0001), as well as upregulated genes, met proto-oncogene (fold change 2.5 ± 0.7, P = .029), and periostin (fold change 7.5 ± 3.4, P = .003). No significant differences in gene expression was found for neurabin 2 (fold change 1.0, P = .99). CONCLUSION: The transcriptional pattern of ethmoid polyps appears to be unique compared with other subsites in the sinonasal cavity of patients with chronic sinusitis. Care must be taken when collecting specimens for molecular studies of the sinonasal tract to differentiate polyp from nonpolyp tissue in chronic sinusitis.


Subject(s)
Ethmoid Sinusitis/genetics , Gene Expression/genetics , Nasal Mucosa/pathology , Nasal Polyps/genetics , Paranasal Sinuses/pathology , Adipokines , Adult , Aged , Aged, 80 and over , Blotting, Western , Carrier Proteins/genetics , Cell Adhesion Molecules/genetics , Chronic Disease , Down-Regulation/genetics , Ethmoid Sinus/pathology , Ethmoid Sinusitis/pathology , Female , Glycoproteins/genetics , Humans , Male , Membrane Transport Proteins , Microfilament Proteins/genetics , Middle Aged , Nasal Polyps/pathology , Nerve Tissue Proteins/genetics , Polymerase Chain Reaction , Prospective Studies , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/genetics , Receptors, Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics , Up-Regulation/genetics
12.
J Biomed Sci ; 16: 67, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19615059

ABSTRACT

Parkinson's disease (PD) is the second most common form of human degenerative disorder. Mutation of parkin is one of the most prevalent causes of autosomal recessive PD. Parkin is an E3 ubiquitin ligase that acts on a variety of substrates, resulting in polyubiquitination and degradation by the proteasome or monoubiquitination and regulation of biological activity. However, the cellular functions of parkin that relate to its pathological involvement in PD are not well understood. Here I show that parkin translocates into nucleus upon DNA damage. Nuclear translocation of parkin appears to be required to promote DNA repair. These findings suggest that DNA damage induces nuclear translocation of parkin leading to the PCNA interaction and possibly other nuclear proteins involved in DNA repair. These results suggest that parkin promotes DNA repair and protects against genotoxicity, and implicate DNA damage as a potential pathogenic mechanism in parkinsonism.


Subject(s)
Active Transport, Cell Nucleus , Brain/metabolism , DNA Damage , Ubiquitin-Protein Ligases/biosynthesis , Adult , Aged , Brain/pathology , Cell Line, Tumor , DNA/radiation effects , DNA Repair , HeLa Cells , Humans , Immunohistochemistry/methods , Models, Biological , Ubiquitin/chemistry , Ultraviolet Rays
13.
Biochem Biophys Res Commun ; 385(3): 434-8, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19470380

ABSTRACT

While mutation of alpha-synuclein is a cause of autosomal-dominant Parkinson's disease (PD), it is still elusive as to how alpha-synuclein is involved in the pathogenesis of PD. Here, we show that dopamine-dependent accumulation of alpha-synuclein in cultured cells results in apoptosis. Furthermore, activation of insulin-like growth factor 1 (IGF-1) pathway can rescue alpha-synuclein toxicity and suppress alpha-synuclein aggregation through the activation of PI3K/Akt pathways. These results suggest the therapeutic potential of IGF-1 pathway in Parkinson disease.


Subject(s)
Apoptosis , Dopamine/metabolism , Insulin-Like Growth Factor I/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Cells, Cultured , Humans , Mutation , Parkinson Disease/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , alpha-Synuclein/genetics
14.
Biochem Biophys Res Commun ; 382(2): 321-5, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19285961

ABSTRACT

Mutation of parkin is one of the most prevalent causes of autosomal recessive Parkinson's disease (PD). Parkin is an E3 ubiquitin ligase that acts on a variety of substrates, resulting in polyubiquitination and degradation by the proteasome or monoubiquitination and regulation of biological activity. However, the cellular functions of parkin that relate to its pathological involvement in PD are not well understood. Here we show that parkin is essential for optimal repair of DNA damage. Parkin-deficient cells exhibit reduced DNA excision repair that can be restored by transfection of wild-type parkin, but not by transfection of a pathological parkin mutant. Parkin also protects against DNA damage-induced cell death, an activity that is largely lost in the pathological mutant. Moreover, parkin interacts with the proliferating cell nuclear antigen (PCNA), a protein that coordinates DNA excision repair. These results suggest that parkin promotes DNA repair and protects against genotoxicity, and implicate DNA damage as a potential pathogenic mechanism in PD.


Subject(s)
DNA Repair , Nuclear Proteins/metabolism , Parkinson Disease/enzymology , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing , Animals , DNA/radiation effects , DNA Damage , DNA Repair/genetics , HeLa Cells , Humans , Mice , Nuclear Proteins/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , Ultraviolet Rays
15.
Nature ; 429(6994): 883-91, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15190254

ABSTRACT

The ageing of the human brain is a cause of cognitive decline in the elderly and the major risk factor for Alzheimer's disease. The time in life when brain ageing begins is undefined. Here we show that transcriptional profiling of the human frontal cortex from individuals ranging from 26 to 106 years of age defines a set of genes with reduced expression after age 40. These genes play central roles in synaptic plasticity, vesicular transport and mitochondrial function. This is followed by induction of stress response, antioxidant and DNA repair genes. DNA damage is markedly increased in the promoters of genes with reduced expression in the aged cortex. Moreover, these gene promoters are selectively damaged by oxidative stress in cultured human neurons, and show reduced base-excision DNA repair. Thus, DNA damage may reduce the expression of selectively vulnerable genes involved in learning, memory and neuronal survival, initiating a programme of brain ageing that starts early in adult life.


Subject(s)
Aging/physiology , Cerebral Cortex/metabolism , DNA Damage , Gene Expression Profiling , Adult , Aged , Aged, 80 and over , Antioxidants/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Survival , Cells, Cultured , Cerebral Cortex/pathology , DNA Repair/genetics , Gene Expression Regulation , Homeostasis , Humans , Learning , Middle Aged , Neuronal Plasticity/genetics , Neurons/metabolism , Neurons/pathology , Oligonucleotide Array Sequence Analysis , Oxidative Stress/genetics , Promoter Regions, Genetic/genetics
16.
Nat Med ; 8(6): 600-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12042811

ABSTRACT

The mechanism by which dopaminergic neurons are selectively lost in Parkinson disease (PD) is unknown. Here we show that accumulation of alpha-synuclein in cultured human dopaminergic neurons results in apoptosis that requires endogenous dopamine production and is mediated by reactive oxygen species. In contrast, alpha-synuclein is not toxic in non-dopaminergic human cortical neurons, but rather exhibits neuroprotective activity. Dopamine-dependent neurotoxicity is mediated by 54 83-kD soluble protein complexes that contain alpha-synuclein and 14-3-3 protein, which are elevated selectively in the substantia nigra in PD. Thus, accumulation of soluble alpha-synuclein protein complexes can render endogenous dopamine toxic, suggesting a potential mechanism for the selectivity of neuronal loss in PD.


Subject(s)
Dopamine/physiology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/physiology , Neurons/physiology , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Parkinson Disease/physiopathology , 14-3-3 Proteins , Apoptosis , Cells, Cultured , Humans , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Neurons/cytology , Parkinson Disease/pathology , Phosphoproteins/genetics , Phosphoproteins/physiology , Substantia Nigra/pathology , Synucleins , Transfection , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/physiology , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...