Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 5530, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797926

ABSTRACT

The adult mammalian inner ear lacks the capacity to divide or regenerate. Damage to inner ear generally leads to permanent hearing loss in humans. Here, we present that reprogramming of the adult inner ear induces renewed proliferation and regeneration of inner ear cell types. Co-activation of cell cycle activator Myc and inner ear progenitor gene Notch1 induces robust proliferation of diverse adult cochlear sensory epithelial cell types. Transient MYC and NOTCH activities enable adult supporting cells to respond to transcription factor Atoh1 and efficiently transdifferentiate into hair cell-like cells. Furthermore, we uncover that mTOR pathway participates in MYC/NOTCH-mediated proliferation and regeneration. These regenerated hair cell-like cells take up the styryl dye FM1-43 and are likely to form connections with adult spiral ganglion neurons, supporting that Myc and Notch1 co-activation is sufficient to reprogram fully mature supporting cells to proliferate and regenerate hair cell-like cells in adult mammalian auditory organs.


Subject(s)
Cell Proliferation/physiology , Cochlea/physiology , Hair Cells, Auditory, Inner/physiology , Regeneration/physiology , Animals , Cell Proliferation/genetics , Cochlea/cytology , Cochlea/metabolism , Ear, Inner/cytology , Ear, Inner/metabolism , Ear, Inner/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Ganglia, Sensory/cytology , Ganglia, Sensory/metabolism , Ganglia, Sensory/physiology , Gene Expression Regulation , Hair Cells, Auditory, Inner/metabolism , Humans , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Regeneration/genetics
2.
Sci Data ; 5: 180128, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30015805

ABSTRACT

Hair cells of the inner ear undergo postnatal development that leads to formation of their sensory organelles, synaptic machinery, and in the case of cochlear outer hair cells, their electromotile mechanism. To examine how the proteome changes over development from postnatal days 0 through 7, we isolated pools of 5000 Pou4f3-Gfp positive or negative cells from the cochlea or utricles; these cell pools were analysed by data-dependent and data-independent acquisition (DDA and DIA) mass spectrometry. DDA data were used to generate spectral libraries, which enabled identification and accurate quantitation of specific proteins using the DIA datasets. DIA measurements were extremely sensitive; we were able to detect proteins present at less than one part in 100,000 from only 312 hair cells. The DDA and DIA datasets will be valuable for accurately quantifying proteins in hair cells and non-hair cells over this developmental window.


Subject(s)
Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/metabolism , Proteome , Animals , Cochlea/cytology , Cochlea/growth & development , Cochlea/metabolism , Mass Spectrometry , Mice
3.
J Cell Biol ; 216(11): 3861-3881, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28899994

ABSTRACT

Control of the dimensions of actin-rich processes like filopodia, lamellipodia, microvilli, and stereocilia requires the coordinated activity of many proteins. Each of these actin structures relies on heterodimeric capping protein (CAPZ), which blocks actin polymerization at barbed ends. Because dimension control of the inner ear's stereocilia is particularly precise, we studied the CAPZB subunit in hair cells. CAPZB, present at ∼100 copies per stereocilium, concentrated at stereocilia tips as hair cell development progressed, similar to the CAPZB-interacting protein TWF2. We deleted Capzb specifically in hair cells using Atoh1-Cre, which eliminated auditory and vestibular function. Capzb-null stereocilia initially developed normally but later shortened and disappeared; surprisingly, stereocilia width decreased concomitantly with length. CAPZB2 expressed by in utero electroporation prevented normal elongation of vestibular stereocilia and irregularly widened them. Together, these results suggest that capping protein participates in stereocilia widening by preventing newly elongating actin filaments from depolymerizing.


Subject(s)
CapZ Actin Capping Protein/metabolism , Hair Cells, Auditory/metabolism , Animals , Auditory Threshold , Behavior, Animal , Brain Stem/metabolism , Brain Stem/physiopathology , CapZ Actin Capping Protein/deficiency , CapZ Actin Capping Protein/genetics , Chick Embryo , Cilia/metabolism , Cilia/ultrastructure , Evoked Potentials, Auditory, Brain Stem , Gene Expression Regulation, Developmental , Genotype , Hair Cells, Auditory/ultrastructure , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Otoacoustic Emissions, Spontaneous , Phenotype , Vestibular Evoked Myogenic Potentials , Vestibule, Labyrinth/metabolism , Vestibule, Labyrinth/physiopathology
4.
Mol Ther ; 25(2): 379-391, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28082074

ABSTRACT

Adeno-associated virus (AAV) is a safe and effective vector for gene therapy for retinal disorders. Gene therapy for hearing disorders is not as advanced, in part because gene delivery to sensory hair cells of the inner ear is inefficient. Although AAV transduces the inner hair cells of the mouse cochlea, outer hair cells remain refractory to transduction. Here, we demonstrate that a vector, exosome-associated AAV (exo-AAV), is a potent carrier of transgenes to all inner ear hair cells. Exo-AAV1-GFP is more efficient than conventional AAV1-GFP, both in mouse cochlear explants in vitro and with direct cochlear injection in vivo. Exo-AAV shows no toxicity in vivo, as assayed by tests of auditory and vestibular function. Finally, exo-AAV1 gene therapy partially rescues hearing in a mouse model of hereditary deafness (lipoma HMGIC fusion partner-like 5/tetraspan membrane protein of hair cell stereocilia [Lhfpl5/Tmhs-/-]). Exo-AAV is a powerful gene delivery system for hair cell research and may be useful for gene therapy for deafness.


Subject(s)
Dependovirus/genetics , Exosomes/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Hair Cells, Auditory, Inner/metabolism , Hearing/genetics , Animals , Cells, Cultured , Dependovirus/classification , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Male , Mice , Mice, Knockout , Phenotype , Transduction, Genetic , Transgenes
5.
Sci Rep ; 5: 18568, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26689366

ABSTRACT

Hereditary Hearing Loss (HHL) is an extremely heterogeneous disorder. Approximately 30 out of 80 known HHL genes are associated with autosomal dominant forms. Here, we identified PSIP1/LEDGF (isoform p75) as a novel strong candidate gene involved in dominant HHL. Using exome sequencing we found a frameshift deletion (c.1554_1555del leading to p.E518Dfs*2) in an Italian pedigree affected by sensorineural mild-to-moderate HHL but also showing a variable eye phenotype (i.e. uveitis, optic neuropathy). This deletion led to a premature stop codon (p.T519X) with truncation of the last 12 amino acids. PSIP1 was recently described as a transcriptional co-activator regulated by miR-135b in vestibular hair cells of the mouse inner ear as well as a possible protector against photoreceptor degeneration. Here, we demonstrate that it is ubiquitously expressed in the mouse inner ear. The PSIP1 mutation is associated with a peculiar audiometric slope toward the high frequencies. These findings indicate that PSIP1 likely plays an important role in HHL.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Genetic Predisposition to Disease , Hearing Loss, Sensorineural/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , DNA Mutational Analysis , Ear, Inner , Exome/genetics , Family , Female , Frameshift Mutation/genetics , Gene Expression Regulation , Humans , Male , Mice , Molecular Sequence Data , Mutation/genetics , Pedigree , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Transcription Factors/chemistry , Transcription Factors/metabolism , Young Adult
6.
Database (Oxford) ; 2015: bav071, 2015.
Article in English | MEDLINE | ID: mdl-26209310

ABSTRACT

The inner ear is a highly specialized mechanosensitive organ responsible for hearing and balance. Its small size and difficulty in harvesting sufficient tissue has hindered the progress of molecular studies. The protein components of mechanotransduction, the molecular biology of inner ear development and the genetic causes of many hereditary hearing and balance disorders remain largely unknown. Inner-ear gene expression data will help illuminate each of these areas. For over a decade, our laboratories and others have generated extensive sets of gene expression data for different cell types in the inner ear using various sample preparation methods and high-throughput genome-wide approaches. To facilitate the study of genes in the inner ear by efficient presentation of the accumulated data and to foster collaboration among investigators, we have developed the Shared Harvard Inner Ear Laboratory Database (SHIELD), an integrated resource that seeks to compile, organize and analyse the genomic, transcriptomic and proteomic knowledge of the inner ear. Five datasets are currently available. These datasets are combined in a relational database that integrates experimental data and annotations relevant to the inner ear. The SHIELD has a searchable web interface with two data retrieval options: viewing the gene pages online or downloading individual datasets as data tables. Each retrieved gene page shows the gene expression data and detailed gene information with hyperlinks to other online databases with up-to-date annotations. Downloadable data tables, for more convenient offline data analysis, are derived from publications and are current as of the time of publication. The SHIELD has made published and some unpublished data freely available to the public with the hope and expectation of accelerating discovery in the molecular biology of balance, hearing and deafness.


Subject(s)
Databases, Genetic , Ear, Inner , Gene Expression Regulation , Genome-Wide Association Study , Genomics , Mechanotransduction, Cellular , Animals , Humans
7.
Hum Mol Genet ; 24(19): 5655-64, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26188009

ABSTRACT

Hearing loss and individual differences in normal hearing both have a substantial genetic basis. Although many new genes contributing to deafness have been identified, very little is known about genes/variants modulating the normal range of hearing ability. To fill this gap, we performed a two-stage meta-analysis on hearing thresholds (tested at 0.25, 0.5, 1, 2, 4, 8 kHz) and on pure-tone averages (low-, medium- and high-frequency thresholds grouped) in several isolated populations from Italy and Central Asia (total N = 2636). Here, we detected two genome-wide significant loci close to PCDH20 and SLC28A3 (top hits: rs78043697, P = 4.71E-10 and rs7032430, P = 2.39E-09, respectively). For both loci, we sought replication in two independent cohorts: B58C from the UK (N = 5892) and FITSA from Finland (N = 270). Both loci were successfully replicated at a nominal level of significance (P < 0.05). In order to confirm our quantitative findings, we carried out RT-PCR and reported RNA-Seq data, which showed that both genes are expressed in mouse inner ear, especially in hair cells, further suggesting them as good candidates for modulatory genes in the auditory system. Sequencing data revealed no functional variants in the coding region of PCDH20 or SLC28A3, suggesting that variation in regulatory sequences may affect expression. Overall, these results contribute to a better understanding of the complex mechanisms underlying human hearing function.


Subject(s)
Cadherins/genetics , Genome-Wide Association Study/methods , Hearing/physiology , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , Asia, Central , Cadherins/metabolism , Deafness/genetics , Genetic Predisposition to Disease , Hair Cells, Auditory, Inner/metabolism , Hearing/genetics , Humans , Italy , Membrane Transport Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Protocadherins , Sequence Analysis, RNA/methods
8.
J Neurosci ; 35(16): 6366-80, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904789

ABSTRACT

Hair cells of the inner ear are essential for hearing and balance. As a consequence, pathogenic variants in genes specifically expressed in hair cells often cause hereditary deafness. Hair cells are few in number and not easily isolated from the adjacent supporting cells, so the biochemistry and molecular biology of hair cells can be difficult to study. To study gene expression in hair cells, we developed a protocol for hair cell isolation by FACS. With nearly pure hair cells and surrounding cells, from cochlea and utricle and from E16 to P7, we performed a comprehensive cell type-specific RNA-Seq study of gene expression during mouse inner ear development. Expression profiling revealed new hair cell genes with distinct expression patterns: some are specific for vestibular hair cells, others for cochlear hair cells, and some are expressed just before or after maturation of mechanosensitivity. We found that many of the known hereditary deafness genes are much more highly expressed in hair cells than surrounding cells, suggesting that genes preferentially expressed in hair cells are good candidates for unknown deafness genes.


Subject(s)
Gene Expression Regulation, Developmental , Hair Cells, Auditory, Inner/metabolism , Animals , Cell Separation , Flow Cytometry , Gene Expression Profiling , Hair Cells, Auditory, Inner/cytology , Mice , Mice, Transgenic , Saccule and Utricle/cytology , Saccule and Utricle/growth & development , Saccule and Utricle/metabolism
9.
Cell Rep ; 10(11): 1811-8, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25772365

ABSTRACT

Hair cells of the inner ear are mechanoreceptors for hearing and balance, and proteins highly enriched in hair cells may have specific roles in the development and maintenance of the mechanotransduction apparatus. We identified XIRP2/mXinß as an enriched protein likely to be essential for hair cells. We found that different isoforms of this protein are expressed and differentially located: short splice forms (also called XEPLIN) are targeted more to stereocilia, whereas two long isoforms containing a XIN-repeat domain are in both stereocilia and cuticular plates. Mice lacking the Xirp2 gene developed normal stereocilia bundles, but these degenerated with time: stereocilia were lost and long membranous protrusions emanated from the nearby apical surfaces. At an ultrastructural level, the paracrystalline actin filaments became disorganized. XIRP2 is apparently involved in the maintenance of actin structures in stereocilia and cuticular plates of hair cells, and perhaps in other organs where it is expressed.


Subject(s)
DNA-Binding Proteins/metabolism , Hair Cells, Auditory/metabolism , LIM Domain Proteins/metabolism , Nuclear Proteins/metabolism , Stereocilia/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , Hair Cells, Auditory/ultrastructure , LIM Domain Proteins/genetics , Mice , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport
10.
J Neurosci ; 33(38): 15086-94, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048839

ABSTRACT

Isl1 is a LIM-homeodomain transcription factor that is critical in the development and differentiation of multiple tissues. In the mouse inner ear, Isl1 is expressed in the prosensory region of otocyst, in young hair cells and supporting cells, and is no longer expressed in postnatal auditory hair cells. To evaluate how continuous Isl1 expression in postnatal hair cells affects hair cell development and cochlear function, we created a transgenic mouse model in which the Pou4f3 promoter drives Isl1 overexpression specifically in hair cells. Isl1 overexpressing hair cells develop normally, as seen by morphology and cochlear functions (auditory brainstem response and otoacoustic emissions). As the mice aged to 17 months, wild-type (WT) controls showed the progressive threshold elevation and outer hair cell loss characteristic of the age-related hearing loss (ARHL) in the background strain (C57BL/6J). In contrast, the Isl1 transgenic mice showed significantly less threshold elevation with survival of hair cells. Further, the Isl1 overexpression protected the ear from noise-induced hearing loss (NIHL): both ABR threshold shifts and hair cell death were significantly reduced when compared with WT littermates. Our model suggests a common mechanism underlying ARHL and NIHL, and provides evidence that hair cell-specific Isl1 expression can promote hair cell survival and therefore minimize the hearing impairment that normally occurs with aging and/or acoustic overexposure.


Subject(s)
Aging , Gene Expression Regulation/physiology , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/pathology , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Acoustic Stimulation , Analysis of Variance , Animals , Cochlea/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hearing Loss, Noise-Induced/metabolism , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otoacoustic Emissions, Spontaneous , RNA, Messenger/metabolism , Rats , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics
11.
J Neurochem ; 103(6): 2651-64, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17961150

ABSTRACT

Acetylcholine is a key neurotransmitter of the inner ear efferent system. In this study, we identify two novel nAChR subunits in the inner ear: α1 and γ, encoded by Chrna1 and Chrng, respectively. In situ hybridization shows that the messages of these two subunits are present in vestibular and cochlear hair cells during early development. Chrna1 and Chrng expression begin at embryonic stage E13.5 in the vestibular system and E17.5 in the organ of Corti. Chrna1 message continues through P7, whereas Chrng is undetectable at post-natal stage P6. The α1 and γ subunits are known as muscle-type nAChR subunits and are surprisingly expressed in hair cells which are sensory-neural cells. We also show that ATOH1/MATH1, a transcription factor essential for hair cell development, directly activates CHRNA1 transcription. Electrophoretic mobility-shift assays and supershift assays showed that ATOH1/E47 heterodimers selectively bind on two E boxes located in the proximal promoter of CHRNA1. Thus, Chrna1 could be the first transcriptional target of ATOH1 in the inner ear. Co-expression in Xenopus oocytes of the α1 subunit does not change the electrophysiological properties of the α9α10 receptor. We suggest that hair cells transiently express α1γ-containing nAChRs in addition to α9α10, and that these may have a role during development of the inner ear innervation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Ear, Inner/metabolism , Hair Cells, Auditory, Inner/metabolism , Receptors, Nicotinic/biosynthesis , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Ear, Inner/embryology , Electrophoretic Mobility Shift Assay , Electrophysiological Phenomena , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/physiology , Luciferases/metabolism , Mice , Molecular Sequence Data , Oocytes/metabolism , Patch-Clamp Techniques , Plasmids/genetics , Pregnancy , RNA/biosynthesis , RNA/genetics , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenopus laevis
12.
FEBS Lett ; 581(24): 4651-6, 2007 Oct 02.
Article in English | MEDLINE | ID: mdl-17826772

ABSTRACT

ATOH1 is a basic Helix-Loop-Helix transcription factor crucial for hair cell (HC) differentiation in the inner ear. In order to identify ATOH1 target genes, we performed a genome-wide expression profiling analysis in cells expressing ATOH1 under the control of a tetracycline-off system and found that HES6 expression is induced by ATOH1. We performed in situ hybridisation and showed that the rise and fall of Hes6 expression closely follow that of Atoh1 in cochlear HC. Moreover, electrophoretic mobility shift assays and luciferase assays show that ATOH1 activates HES6 transcription through binding to three clustered E boxes of its promoter.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Hair Cells, Auditory/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Conserved Sequence , Gene Expression Profiling , Humans , Mice , Molecular Sequence Data , Protein Binding , Repressor Proteins/genetics , Sequence Alignment , Transcription, Genetic/genetics
13.
Am J Med Genet A ; 128A(1): 29-32, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15211652

ABSTRACT

Desbuquois dysplasia is a rare chondrodysplasia of autosomal recessive inheritance characterized by short stature, joint laxity, facial anomalies, a "Swedish key" appearance of the proximal femur, and advanced carpal and tarsal bone age. Patients with Desbuquois dysplasia can be divided in two groups, depending on whether hand changes include an extra ossification center distal to the second metacarpal and whether phalangeal dislocations are present or absent. We have recently reported linkage of a Desbuquois dysplasia gene to 17q25.3 in a group of patients with typical hand abnormalities. Here, we report on the exclusion of the 17q25.3 locus in three inbred Desbuquois families originated from Turkey, Asia, and Morocco without typical hand abnormalities. Microsatellite DNA markers from the 17q25.3 region were used at an average spacing of 2 cM, and the three affected individuals from families 1 to 3 were heterozygous for the 17q25.3 region. These results allow us to exclude this region as the locus in Desbuquois families with no hand anomalies and demonstrate genetic heterogeneity. Ongoing studies will hopefully lead to the identification of the responsible genes.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 17 , Genetic Heterogeneity , Genetic Markers , Osteochondrodysplasias/genetics , Adolescent , Body Height , Child , Chromosome Mapping , Face/abnormalities , Female , Femur/abnormalities , Hand Deformities/genetics , Humans , Inbreeding , Infant, Newborn , Joint Instability/genetics , Male , Microsatellite Repeats , Osteochondrodysplasias/pathology , Pedigree
14.
Am J Hum Genet ; 74(2): 298-305, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14740318

ABSTRACT

Stuve-Wiedemann syndrome (SWS) is a severe autosomal recessive condition characterized by bowing of the long bones, with cortical thickening, flared metaphyses with coarsened trabecular pattern, camptodactyly, respiratory distress, feeding difficulties, and hyperthermic episodes responsible for early lethality. Clinical overlap with Schwartz-Jampel type 2 syndrome (SJS2) has suggested that SWS and SJS2 could be allelic disorders. Through studying a series of 19 families with SWS/SJS2, we have mapped the disease gene to chromosome 5p13.1 at locus D5S418 (Zmax=10.66 at theta =0) and have identified null mutations in the leukemia inhibitory factor receptor (LIFR or gp190 chain) gene. A total of 14 distinct mutations were identified in the 19 families. An identical frameshift insertion (653_654insT) was identified in families from the United Arab Emirates, suggesting a founder effect in that region. It is interesting that 12/14 mutations predicted premature termination of translation. Functional studies indicated that these mutations alter the stability of LIFR messenger RNA transcripts, resulting in the absence of the LIFR protein and in the impairment of the JAK/STAT3 signaling pathway in patient cells. We conclude, therefore, that SWS and SJS2 represent a single clinically and genetically homogeneous condition due to null mutations in the LIFR gene on chromosome 5p13.


Subject(s)
Mutation , Osteochondrodysplasias/genetics , Receptors, Cytokine/genetics , Base Sequence , Child , Chromosomes, Human, Pair 5 , DNA Primers , Genetic Linkage , Humans , Immunohistochemistry , Leukemia Inhibitory Factor Receptor alpha Subunit , Molecular Sequence Data , Receptors, OSM-LIF
SELECTION OF CITATIONS
SEARCH DETAIL
...