Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Development ; 146(20)2019 10 04.
Article in English | MEDLINE | ID: mdl-31540914

ABSTRACT

The transcription factor scleraxis (Scx) is required for tendon development; however, the function of Scx is not fully understood. Although Scx is expressed by all tendon progenitors and cells, only long tendons are disrupted in the Scx-/- mutant; short tendons appear normal and the ability of muscle to attach to skeleton is not affected. We recently demonstrated that long tendons are formed in two stages: first, by muscle anchoring to skeleton via a short tendon anlage; and second, by rapid elongation of the tendon in parallel with skeletal growth. Through lineage tracing, we extend these observations to all long tendons and show that tendon elongation is fueled by recruitment of new mesenchymal progenitors. Conditional loss of Scx in mesenchymal progenitors did not affect the first stage of anchoring; however, new cells were not recruited during elongation and long tendon formation was impaired. Interestingly, for tenocyte recruitment, Scx expression was required only in the recruited cells and not in the recruiting tendon. The phenotype of Scx mutants can thus be understood as a failure of tendon cell recruitment during tendon elongation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Tendons/cytology , Tendons/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Mice , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism
2.
Nucleic Acids Res ; 48(9): 5065-5080, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32249312

ABSTRACT

Disabling hearing loss impacts ∼466 million individuals worldwide with 34 million children affected. Gene and pharmacotherapeutic strategies to rescue auditory function in mouse models of human deafness are most effective when administered before hearing onset, after which therapeutic efficacy is significantly diminished or lost. We hypothesize that preemptive correction of a mutation in the fetal inner ear prior to maturation of the sensory epithelium will optimally restore sensory function. We previously demonstrated that transuterine microinjection of a splice-switching antisense oligonucleotide (ASO) into the amniotic cavity immediately surrounding the embryo on embryonic day 13-13.5 (E13-13.5) corrected pre-mRNA splicing in the juvenile Usher syndrome type 1c (Ush1c) mouse mutant. Here, we show that this strategy only marginally rescues hearing and partially rescues vestibular function. To improve therapeutic outcomes, we microinjected ASO directly into the E12.5 inner ear. A single intra-otic dose of ASO corrects harmonin RNA splicing, restores harmonin protein expression in sensory hair cell bundles, prevents hair cell loss, improves hearing sensitivity, and ameliorates vestibular dysfunction. Improvements in auditory and vestibular function were sustained well into adulthood. Our results demonstrate that an ASO pharmacotherapeutic administered to a developing organ system in utero preemptively corrects pre-mRNA splicing to abrogate the disease phenotype.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Deafness/congenital , Deafness/drug therapy , Oligonucleotides, Antisense/therapeutic use , Vestibule, Labyrinth/physiopathology , Amnion , Animals , Auditory Threshold/drug effects , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Deafness/genetics , Deafness/physiopathology , Ear, Inner/drug effects , Ear, Inner/metabolism , Fetus , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Mice , Microinjections , Mutation , Oligonucleotides, Antisense/administration & dosage , RNA Splicing/drug effects , Vestibule, Labyrinth/drug effects
3.
Mol Ther ; 32(4): 859-860, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38522426
4.
Nucleic Acids Res ; 44(20): 9519-9529, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27683224

ABSTRACT

Congenital diseases account for a large portion of pediatric illness. Prenatal screening and diagnosis permit early detection of many genetic diseases. Fetal therapeutic strategies to manage disease processes in utero represent a powerful new approach for clinical care. A safe and effective fetal pharmacotherapy designed to modulate gene expression ideally would avoid direct mechanical engagement of the fetus and present an external reservoir of drug. The amniotic cavity surrounding the fetus could serve as an ideal drug reservoir. Antisense oligonucleotides (ASOs) are an established tool for the therapeutic modulation of gene expression. We hypothesize that ASOs administered to the amniotic cavity will gain entry to the fetus and modulate gene expression. Here, we show that an ASO targeting MALAT1 RNA, delivered by transuterine microinjection into the mouse amniotic cavity at embryonic day 13-13.5, reduces target RNA expression for up to 4 weeks after birth. A similarly delivered ASO targeting a causal splice site mutation for Usher syndrome corrects gene expression in the inner ear, a therapeutically relevant target tissue. We conclude that intra-amniotic delivery of ASOs is well tolerated and produces a sustained effect on postnatal gene expression. Transuterine delivery of ASOs is an innovative platform for developing fetal therapeutics to efficaciously treat congenital disease.


Subject(s)
Amnion/metabolism , Gene Expression Regulation , Microinjections , Oligonucleotides, Antisense/administration & dosage , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cytoskeletal Proteins , Female , Fetus , Gene Expression , Male , Mice , Organ Specificity/genetics , Pregnancy , RNA, Long Noncoding/genetics , RNA, Messenger/genetics
5.
Biophys J ; 110(2): 493-502, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26789771

ABSTRACT

Normal hearing in mammals depends on sound amplification by outer hair cells (OHCs) presumably by their somatic motility and force production. However, the role of OHC force production in cochlear amplification and frequency tuning are not yet fully understood. Currently, available OHC manipulation techniques for physiological or clinical studies are limited by their invasive nature, lack of precision, and poor temporal-spatial resolution. To overcome these limitations, we explored an optogenetic approach based on channelrhodopsin 2 (ChR-2), a direct light-activated nonselective cation channel originally discovered in Chlamydomonas reinhardtii. Three approaches were compared: 1) adeno-associated virus-mediated in utero transfer of the ChR-2 gene into the developing murine otocyst, 2) expression of ChR-2(H134R) in an auditory cell line (HEI-OC1), and 3) expression of ChR-2 in the OHCs of a mouse line carrying a ChR-2 conditional allele. Whole cell recording showed that blue light (470 nm) elicited the typical nonselective cation current of ChR-2 with reversal potential around zero in both mouse OHCs and HEI-OC1 cells and generated depolarization in both cell types. In addition, pulsed light stimulation (10 Hz) elicited a 1:1 repetitive depolarization and ChR-2 currents in mouse OHCs and HEI-OC1 cells, respectively. The time constant of depolarization in OHCs, 1.45 ms, is 10 times faster than HEI-OC1 cells, which allowed light stimulation up to rates of 10/s to elicit corresponding membrane potential changes. Our study demonstrates that ChR-2 can successfully be expressed in mouse OHCs and HEI-OC1 cells and that these present a typical light-sensitive current and depolarization. However, the amount of ChR-2 current induced in our in vivo experiments was insufficient to result in measurable cochlear effects.


Subject(s)
Hair Cells, Auditory, Outer/metabolism , Optogenetics/methods , Action Potentials , Animals , Cell Line , Channelrhodopsins , Hair Cells, Auditory, Outer/physiology , Mice
6.
Nature ; 455(7212): 537-41, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18754012

ABSTRACT

Sensory hair cells in the mammalian cochlea convert mechanical stimuli into electrical impulses that subserve audition. Loss of hair cells and their innervating neurons is the most frequent cause of hearing impairment. Atonal homologue 1 (encoded by Atoh1, also known as Math1) is a basic helix-loop-helix transcription factor required for hair-cell development, and its misexpression in vitro and in vivo generates hair-cell-like cells. Atoh1-based gene therapy to ameliorate auditory and vestibular dysfunction has been proposed. However, the biophysical properties of putative hair cells induced by Atoh1 misexpression have not been characterized. Here we show that in utero gene transfer of Atoh1 produces functional supernumerary hair cells in the mouse cochlea. The induced hair cells display stereociliary bundles, attract neuronal processes and express the ribbon synapse marker carboxy-terminal binding protein 2 (refs 12,13). Moreover, the hair cells are capable of mechanoelectrical transduction and show basolateral conductances with age-appropriate specializations. Our results demonstrate that manipulation of cell fate by transcription factor misexpression produces functional sensory cells in the postnatal mammalian cochlea. We expect that our in utero gene transfer paradigm will enable the design and validation of gene therapies to ameliorate hearing loss in mouse models of human deafness.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cochlea/cytology , Cochlea/metabolism , Hair Cells, Auditory/physiology , Transfection , Uterus , Animals , Cochlea/embryology , Cochlea/growth & development , Cochlea/innervation , Deafness/genetics , Deafness/therapy , Disease Models, Animal , Dyneins/metabolism , Electric Conductivity , Female , Genetic Therapy , Hair Cells, Auditory/cytology , Humans , Mechanotransduction, Cellular , Mice , Myosin VIIa , Myosins/metabolism , Pregnancy , Synapses/metabolism
7.
J Neurosci ; 31(13): 4886-95, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21451027

ABSTRACT

Cochlear inner hair cells (IHCs) use Ca(2+)-dependent exocytosis of glutamate to signal sound information. Otoferlin (Otof), a C(2) domain protein essential for IHC exocytosis and hearing, may serve as a Ca(2+) sensor in vesicle fusion in IHCs that seem to lack the classical neuronal Ca(2+) sensors synaptotagmin 1 (Syt1) and Syt2. Support for the Ca(2+) sensor of fusion hypothesis for otoferlin function comes from biochemical experiments, but additional roles in late exocytosis upstream of fusion have been indicated by physiological studies. Here, we tested the functional equivalence of otoferlin and Syt1 in three neurosecretory model systems: auditory IHCs, adrenal chromaffin cells, and hippocampal neurons. Long-term and short-term ectopic expression of Syt1 in IHCs of Otof (-/-) mice by viral gene transfer in the embryonic inner ear and organotypic culture failed to rescue their Ca(2+) influx-triggered exocytosis. Conversely, virally mediated overexpression of otoferlin did not restore phasic exocytosis in Syt1-deficient chromaffin cells or neurons but enhanced asynchronous release in the latter. We further tested exocytosis in Otof (-/-) hippocampal neurons and in Syt1(-/-) IHCs but found no deficits in vesicle fusion. Expression analysis of different synaptotagmin isoforms indicated that Syt1 and Syt2 are absent from mature IHCs. Our data argue against a simple functional equivalence of the two C(2) domain proteins in exocytosis of IHC ribbon synapses, chromaffin cells, and hippocampal synapses.


Subject(s)
Exocytosis/physiology , Membrane Proteins/physiology , Synaptotagmin I/physiology , Acoustic Stimulation/methods , Animals , Animals, Newborn , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Exocytosis/genetics , Hippocampus/cytology , Hippocampus/physiology , Membrane Fusion/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Knockout , Neural Inhibition/genetics , Neurons/metabolism , Organ Culture Techniques , Synapses/genetics , Synapses/physiology , Synaptotagmin I/deficiency , Synaptotagmin I/genetics
8.
Sci Rep ; 12(1): 10036, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35710827

ABSTRACT

Mutations in the MYO7A gene lead to Usher syndrome type 1B (USH1B), a disease characterized by congenital deafness, vision loss, and balance impairment. To create a nonhuman primate (NHP) USH1B model, CRISPR/Cas9 was used to disrupt MYO7A in rhesus macaque zygotes. The targeting efficiency of Cas9 mRNA and hybridized crRNA-tracrRNA (hyb-gRNA) was compared to Cas9 nuclease (Nuc) protein and synthetic single guide (sg)RNAs. Nuc/sgRNA injection led to higher editing efficiencies relative to mRNA/hyb-gRNAs. Mutations were assessed by preimplantation genetic testing (PGT) and those with the desired mutations were transferred into surrogates. A pregnancy was established from an embryo where 92.1% of the PGT sequencing reads possessed a single G insertion that leads to a premature stop codon. Analysis of single peripheral blood leukocytes from the infant revealed that half the cells possessed the homozygous single base insertion and the remaining cells had the wild-type MYO7A sequence. The infant showed sensitive auditory thresholds beginning at 3 months. Although further optimization is needed, our studies demonstrate that it is feasible to use CRISPR technologies for creating NHP models of human diseases.


Subject(s)
Usher Syndromes , Animals , Humans , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing , Macaca mulatta/genetics , Macaca mulatta/metabolism , RNA, Messenger , Usher Syndromes/genetics , RNA, Small Untranslated/metabolism
9.
Hear Res ; 394: 107931, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32173115

ABSTRACT

Disabling hearing loss is expected to affect over 900 million people worldwide by 2050. The World Health Organization estimates that the annual economic impact of hearing loss globally is US$ 750 billion. The inability to hear may complicate effective interpersonal communication and negatively impact personal and professional relationships. Recent advances in the genetic diagnosis of inner ear disease have keenly focused attention on strategies to restore hearing and balance in individuals with defined gene mutations. Mouse models of human hearing loss serve as the primary approach to test gene therapies and pharmacotherapies. The goal of this review is to articulate the rationale for fetal gene therapy and pharmacotherapy to treat congenital hearing loss and vestibular dysfunction. The differential onset of hearing in mice and humans suggests that a prenatal window of therapeutic efficacy in humans may be optimal to restore sensory function. Mouse studies demonstrating the utility of early fetal intervention in the inner ear show promise. We focus on the modulation of gene expression through two strategies that have successfully treated deafness in animal models and have had clinical success for other conditions in humans: gene replacement and antisense oligonucleotide-mediated modulation of gene expression. The recent establishment of effective therapies targeting the juvenile and adult mouse provide informative counterexamples where intervention in the maturing and fully functional mouse inner ear may be effective. Distillation of the current literature leads to the conclusion that novel therapeutic strategies to treat genetic deafness and imbalance will soon translate to clinical trials.


Subject(s)
Genetic Therapy , Hearing Loss , Animals , Deafness , Ear, Inner , Hearing Loss/genetics , Hearing Loss/therapy , Hearing Loss, Sensorineural , Mice
10.
J Assoc Res Otolaryngol ; 21(3): 225-242, 2020 06.
Article in English | MEDLINE | ID: mdl-32648066

ABSTRACT

Auditory brainstem responses (ABRs) require averaging responses to hundreds or thousands of repetitions of a stimulus (e.g., tone pip) to obtain a measurable evoked response at the scalp. Fast repetition rates lead to changes in ABR amplitude and latency due to adaptation. To minimize the effect of adaptation, stimulus rates are sometimes as low as 10 to 13.3 stimuli per second, requiring long acquisition times. The trade-off between reducing acquisition time and minimizing the effect of adaptation on ABRs is an especially important consideration for studies of cochlear synaptopathy, which use the amplitude of short latency responses (wave 1) to assess auditory nerve survival. It has been proposed that adaptation during ABR acquisition can be reduced by interleaving tones at different frequencies, rather than testing each frequency serially. With careful ordering of frequencies and levels in the stimulus train, adaptation in the auditory nerve can be minimized, thereby permitting an increase in the rate at which tone bursts are presented. However, widespread adoption of this stimulus design has been hindered by lack of available software. Here, we develop and validate an interleaved stimulus design to optimize the rate of ABR measurement while minimizing adaptation. We implement this method in an open-source data acquisition software tool that permits either serial or interleaved ABR measurements. The open-source software library, psiexperiment, is compatible with widely used ABR hardware. Consistent with previous studies, careful design of an interleaved stimulus train can reduce ABR acquisition time by more than half, with minimal effect on ABR thresholds and wave 1 latency, while improving measures of wave 1 amplitude.


Subject(s)
Auditory Perception/physiology , Electrophysiology/methods , Evoked Potentials, Auditory, Brain Stem , Software , Animals , Ferrets , Gerbillinae , Macaca mulatta , Mice
11.
Elife ; 92020 01 21.
Article in English | MEDLINE | ID: mdl-31961320

ABSTRACT

Studies of cell fate focus on specification, but little is known about maintenance of the differentiated state. In this study, we find that the mouse tendon cell fate requires continuous maintenance in vivo and identify an essential role for TGFß signaling in maintenance of the tendon cell fate. To examine the role of TGFß signaling in tenocyte function the TGFß type II receptor (Tgfbr2) was targeted in the Scleraxis-expressing cell lineage using the ScxCre deletor. Tendon development was not disrupted in mutant embryos, but shortly after birth tenocytes lost differentiation markers and reverted to a more stem/progenitor state. Viral reintroduction of Tgfbr2 to mutants prevented and even rescued tenocyte dedifferentiation suggesting a continuous and cell autonomous role for TGFß signaling in cell fate maintenance. These results uncover the critical importance of molecular pathways that maintain the differentiated cell fate and a key role for TGFß signaling in these processes.


Subject(s)
Receptor, Transforming Growth Factor-beta Type II/metabolism , Tendons/cytology , Transforming Growth Factor beta/metabolism , Animals , Cell Dedifferentiation , Cell Lineage , Gene Expression Regulation , Mice , Mutation , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tendons/metabolism , Tenocytes/cytology , Tenocytes/metabolism
12.
PLoS One ; 15(1): e0227676, 2020.
Article in English | MEDLINE | ID: mdl-31935257

ABSTRACT

Zika virus infection during pregnancy is associated with miscarriage and with a broad spectrum of fetal and neonatal developmental abnormalities collectively known as congenital Zika syndrome (CZS). Symptomology of CZS includes malformations of the brain and skull, neurodevelopmental delay, seizures, joint contractures, hearing loss and visual impairment. Previous studies of Zika virus in pregnant rhesus macaques (Macaca mulatta) have described injury to the developing fetus and pregnancy loss, but neonatal outcomes following fetal Zika virus exposure have yet to be characterized in nonhuman primates. Herein we describe the presentation of rhesus macaque neonates with a spectrum of clinical outcomes, including one infant with CZS-like symptoms including cardiomyopathy, motor delay and seizure activity following maternal infection with Zika virus during the first trimester of pregnancy. Further characterization of this neonatal nonhuman primate model of gestational Zika virus infection will provide opportunities to evaluate the efficacy of pre- and postnatal therapeutics for gestational Zika virus infection and CZS.


Subject(s)
Disease Models, Animal , Zika Virus Infection/veterinary , Zika Virus/pathogenicity , Animals , Cardiomyopathies/virology , Female , Fetus/virology , Macaca mulatta , Microcephaly/virology , Pregnancy , Pregnancy Complications, Infectious/veterinary , Pregnancy Complications, Infectious/virology , Pregnancy Trimester, First , Seizures/virology , Zika Virus Infection/virology
13.
Methods Mol Biol ; 493: 125-39, 2009.
Article in English | MEDLINE | ID: mdl-18839345

ABSTRACT

The mammalian inner ear forms from a thickened patch of head ectoderm called the otic placode. The placodal ectoderm invaginates to form a cup whose edges cinch together to establish a fluid-filled sac called the otic vesicle or otocyst. The progenitor cells lining the otocyst lumen will give rise to sensory and non-sensory cells of the inner ear. These formative stages of inner ear development are initiated during the first week of postimplantation embryonic development in the mouse. The inaccessibility of the inner ear in utero has hampered efforts to gain insight into the molecular mechanisms regulating essential developmental processes. An experimental embryological method to misexpress genes in the developing mammalian inner ear is presented. Expression plasmid encoding a gene of interest is microinjected through the uterine wall into the lumen of the otocyst and electroporated into otic epithelial progenitor cells. Downstream analysis of the transfected embryonic or postnatal inner ear is then conducted to gain insight into gene function.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , Electroporation/methods , Transfection/methods , Animals , Ear, Inner/cytology , Female , Gene Expression Regulation, Developmental , Mice , Pregnancy
14.
Front Mol Neurosci ; 11: 300, 2018.
Article in English | MEDLINE | ID: mdl-30210291

ABSTRACT

Therapeutic strategies to restore hearing and balance in mouse models of inner ear disease aim to rescue sensory function by gene replacement, augmentation, knock down or knock out. Modalities to achieve therapeutic effects have utilized virus-mediated transfer of wild type genes and small interfering ribonucleic acids; systemic and focal administration of antisense oligonucleotides (ASO) and designer small molecules; and lipid-mediated transfer of Cas 9 ribonucleoprotein (RNP) complexes. This work has established that gene or drug administration to the structurally and functionally immature, early neonatal mouse inner ear prior to hearing onset is a prerequisite for the most robust therapeutic responses. These observations may have significant implications for translating mouse inner ear gene therapies to patients. The human fetus hears by gestational week 19, suggesting that a corollary window of therapeutic efficacy closes early in the second trimester of pregnancy. We hypothesize that fetal therapeutics deployed prior to hearing onset may be the most effective approach to preemptively manage genetic mutations that cause deafness and vestibular dysfunction. We assert that gene therapy studies in higher vertebrate model systems with fetal hearing onset and a comparable acoustic range and sensitivity to that of humans are an essential step to safely and effectively translate murine gene therapies to the clinic.

15.
Methods Mol Biol ; 1427: 43-57, 2016.
Article in English | MEDLINE | ID: mdl-27259920

ABSTRACT

There is keen interest to define gene therapies aimed at restoration of auditory and vestibular function in the diseased or damaged mammalian inner ear. A persistent limitation of regenerative medical strategies that seek to correct or modify gene expression in the sensory epithelia of the inner ear involves efficacious delivery of a therapeutic genetic construct. Our approach is to define methodologies that enable fetal gene transfer to the developing mammalian inner ear in an effort to correct defective gene expression during formation of the sensory epithelia or during early postnatal life. Conceptually, the goal is to atraumatically introduce the genetic construct into the otocyst-staged mouse inner ear and transfect otic progenitors that give rise to sensory hair cells and supporting cells. Our long-term goal is to define therapeutic interventions for congenital deafness and balance disorders with the expectation that the approach may also be exploited for therapeutic intervention postnatally.In the inaugural volume of this series, we introduced electroporation-mediated gene transfer to the developing mouse inner ear that encompassed our mouse survival surgery and transuterine microinjection protocols (Brigande et al., Methods Mol Biol 493:125-139, 2009). In this chapter, we first briefly update our use of sodium pentobarbital anesthesia, our preferred anesthetic for mouse ventral laparotomy, in light of its rapidly escalating cost. Next, we define a rapid, cost-effective method to produce recombinant adeno-associated virus (rAAV) for efficient gene transfer to the developing mouse inner ear. Our immediate goal is to provide a genetic toolkit that will permit the definition and validation of gene therapies in mouse models of human deafness and balance disorders.


Subject(s)
Deafness/therapy , Dependovirus/genetics , Ear, Inner/embryology , Genetic Engineering/economics , Adjuvants, Anesthesia/administration & dosage , Animals , Deafness/genetics , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy , HEK293 Cells , Humans , Mice , Pentobarbital/administration & dosage
16.
Matrix Biol ; 32(1): 39-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23186870

ABSTRACT

Prolyl 3-hydroxylase1 (P3H1) is a collagen modifying enzyme which hydroxylates certain prolines in the Xaa position of conventional GlyXaaYaa triple helical sequence. Recent investigations have revealed that mutations in the LEPRE1 (gene encoding for P3H1) cause severe osteogenesis imperfecta (OI) in humans. Similarly LEPRE1 knockout mice display an OI-like phenotype. Significant hearing loss is a common problem for people with osteogenesis imperfecta. Here we report that hearing of the P3H1 null mice is substantially affected. Auditory brainstem responses (ABRs) of the P3H1 null mice show an average increase of 20-30 dB in auditory thresholds. Three dimensional reconstructions of the mutant middle ear bones by Micro-scale X-ray computed tomography (Micro-CT) demonstrate abnormal morphology of the incudostapedial and incudomalleal joints. We establish the LEPRE1 knockout mouse as a valuable model system to investigate the mechanism of hearing loss in recessive OI.


Subject(s)
Ear Ossicles/abnormalities , Hearing Loss/genetics , Joints/abnormalities , Membrane Glycoproteins/genetics , Osteogenesis Imperfecta/genetics , Procollagen-Proline Dioxygenase/deficiency , Proteoglycans/genetics , Animals , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem/physiology , Genes, Recessive/genetics , Hearing Loss/enzymology , Mice , Mice, Knockout , Osteogenesis Imperfecta/enzymology , X-Ray Microtomography
17.
PLoS One ; 8(7): e69314, 2013.
Article in English | MEDLINE | ID: mdl-23935981

ABSTRACT

The mammalian inner ear subserves the special senses of hearing and balance. The auditory and vestibular sensory epithelia consist of mechanically sensitive hair cells and associated supporting cells. Hearing loss and balance dysfunction are most frequently caused by compromise of hair cells and/or their innervating neurons. The development of gene- and cell-based therapeutics will benefit from a thorough understanding of the molecular basis of patterning and cell fate specification in the mammalian inner ear. This includes analyses of cell lineages and cell dispersals across anatomical boundaries (such as sensory versus nonsensory territories). The goal of this study was to conduct retroviral lineage analysis of the embryonic day 11.5(E11.5) mouse otic vesicle. A replication-defective retrovirus encoding human placental alkaline phosphatase (PLAP) and a variable 24-bp oligonucleotide tag was microinjected into the E11.5 mouse otocyst. PLAP-positive cells were microdissected from cryostat sections of the postnatal inner ear and subjected to nested PCR. PLAP-positive cells sharing the same sequence tag were assumed to have arisen from a common progenitor and are clonally related. Thirty five multicellular clones consisting of an average of 3.4 cells per clone were identified in the auditory and vestibular sensory epithelia, ganglia, spiral limbus, and stria vascularis. Vestibular hair cells in the posterior crista were related to one another, their supporting cells, and nonsensory epithelial cells lining the ampulla. In the organ of Corti, outer hair cells were related to a supporting cell type and were tightly clustered. By contrast, spiral ganglion neurons, interdental cells, and Claudius' cells were related to cells of the same type and could be dispersed over hundreds of microns. These data contribute new information about the developmental potential of mammalian otic precursors in vivo.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Hair Cells, Auditory/cytology , Hair Cells, Vestibular/cytology , Otolithic Membrane/cytology , Spiral Ganglion/cytology , Stria Vascularis/cytology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Animals, Newborn , Embryo, Mammalian , Epithelial Cells/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Genes, Reporter , Genetic Vectors , Hair Cells, Auditory/metabolism , Hair Cells, Vestibular/metabolism , Humans , Injections , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Microinjections , Morphogenesis/genetics , Oligonucleotides/genetics , Otolithic Membrane/metabolism , Pregnancy , Retroviridae/genetics , Spiral Ganglion/metabolism , Stria Vascularis/metabolism , Uterus
18.
Dev Cell ; 26(5): 544-51, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24044893

ABSTRACT

The muscles that govern hand motion are composed of extrinsic muscles that reside within the forearm and intrinsic muscles that reside within the hand. We find that the extrinsic muscles of the flexor digitorum superficialis (FDS) first differentiate as intrinsic muscles within the hand and then relocate as myofibers to their final position in the arm. This remarkable translocation of differentiated myofibers across a joint is dependent on muscle contraction and muscle-tendon attachment. Interestingly, the intrinsic flexor digitorum brevis (FDB) muscles of the foot are identical to the FDS in tendon pattern and delayed developmental timing but undergo limited muscle translocation, providing strong support for evolutionary homology between the FDS and FDB muscles. We propose that the intrinsic FDB pattern represents the original tetrapod limb and that translocation of the muscles to form the FDS is a mammalian evolutionary addition.


Subject(s)
Forelimb/growth & development , Movement/physiology , Muscles/physiology , Tendons/growth & development , Animals , Foot/anatomy & histology , Foot/physiology , Forelimb/anatomy & histology , Hindlimb/anatomy & histology , Hindlimb/growth & development , Humans , Mice , Muscle Contraction/physiology , Tendons/anatomy & histology
19.
Nat Biotechnol ; 35(3): 216-218, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28267741
20.
J Vis Exp ; (64)2012 Jun 30.
Article in English | MEDLINE | ID: mdl-22781586

ABSTRACT

The mammalian inner ear has 6 distinct sensory epithelia: 3 cristae in the ampullae of the semicircular canals; maculae in the utricle and saccule; and the organ of Corti in the coiled cochlea. The cristae and maculae contain vestibular hair cells that transduce mechanical stimuli to subserve the special sense of balance, while auditory hair cells in the organ of Corti are the primary transducers for hearing. Cell fate specification in these sensory epithelia and morphogenesis of the semicircular canals and cochlea take place during the second week of gestation in the mouse and are largely completed before birth. Developmental studies of the mouse inner ear are routinely conducted by harvesting transgenic embryos at different embryonic or postnatal stages to gain insight into the molecular basis of cellular and/or morphological phenotypes. We hypothesize that gene transfer to the developing mouse inner ear in utero in the context of gain- and loss-of-function studies represents a complimentary approach to traditional mouse transgenesis for the interrogation of the genetic mechanisms underlying mammalian inner ear development(6). The experimental paradigm to conduct gene misexpression studies in the developing mouse inner ear demonstrated here resolves into three general steps: 1) ventral laparotomy; 2) transuterine microinjection; and 3) in vivo electroporation. Ventral laparotomy is a mouse survival surgical technique that permits externalization of the uterus to gain experimental access to the implanted embryos. Transuterine microinjection is the use of beveled, glass capillary micropipettes to introduce expression plasmid into the lumen of the otic vesicle or otocyst. In vivo electroporation is the application of square wave, direct current pulses to drive expression plasmid into progenitor cells. We previously described this electroporation-based gene transfer technique and included detailed notes on each step of the protocol(11). Mouse experimental embryological techniques can be difficult to learn from prose and still images alone. In the present work, we demonstrate the 3 steps in the gene transfer procedure. Most critically, we deploy digital video microscopy to show precisely how to: 1) identify embryo orientation in utero; 2) reorient embryos for targeting injections to the otocyst; 3) microinject DNA mixed with tracer dye solution into the otocyst at embryonic days 11.5 and 12.5; 4) electroporate the injected otocyst; and 5) label electroporated embryos for postnatal selection at birth. We provide representative examples of successfully transfected inner ears; a pictorial guide to the most common causes of otocyst mistargeting; discuss how to avoid common methodological errors; and present guidelines for writing an in utero gene transfer animal care protocol.


Subject(s)
Ear, Inner/physiology , Electroporation/methods , Gene Transfer Techniques , Animals , Cochlea/embryology , Cochlea/physiology , Ear, Inner/embryology , Female , Mice , Microinjections , Microscopy, Video , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL