Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Emerg Nurs ; 50(3): 381-391.e2, 2024 May.
Article in English | MEDLINE | ID: mdl-38506784

ABSTRACT

INTRODUCTION: Freestanding emergency departments (FSEDs) are emergency facilities not connected to inpatient services. The percentage of FSEDs of all EDs grew from 1% in 2001 to 12% in 2017, making FSEDs a substantial subset of US emergency care. The purpose of this study was to describe the individual attributes and environmental conditions of registered nurses working in FSEDs in the US. METHODS: A quantitative descriptive exploratory design with cross-sectional survey methodology. RESULTS: A total of 364 emergency nurses responded to the survey. Most reported their FSED was open 24 hours/day (99.5%), with board-certified emergency physicians onsite (91.5%) and a mean of 3.6 RNs working per shift. Resources immediately available in more than 50% of FSEDs included laboratory and imaging services, and in fewer than 30% of FSEDs included behavioral health care, MRI, obstetric care, orthopedic care, neurologic care, and surgical consult care. Respiratory therapy was reported by 39.6% of respondents as being immediately available. A significant minority of respondents expressed concerns about adequacy of resources and training and the effect on patient care in both survey (30% of respondents) and open-ended questions (42.5% of respondents). DISCUSSION: The practice environment of emergency nurses in FSEDs was reported as having positive elements; however, a substantial subpopulation reported serious concerns. FSEDs adhere to some of the standards put forward by the American College of Emergency Physicians, with notable exceptions in the areas of staffing RNs, staffing ancillary staff, and availability of some resources.


Subject(s)
Emergency Nursing , Emergency Service, Hospital , Humans , Emergency Service, Hospital/statistics & numerical data , United States , Cross-Sectional Studies , Emergency Nursing/statistics & numerical data , Female , Male , Adult , Surveys and Questionnaires , Middle Aged , Nursing Staff, Hospital/statistics & numerical data
2.
Mol Psychiatry ; 27(5): 2492-2501, 2022 05.
Article in English | MEDLINE | ID: mdl-35296810

ABSTRACT

The global crisis of opioid overdose fatalities has led to an urgent search to discover the neurobiological mechanisms of opioid use disorder (OUD). A driving force for OUD is the dysphoric and emotionally painful state (hyperkatifeia) that is produced during acute and protracted opioid withdrawal. Here, we explored a mechanistic role for extrahypothalamic stress systems in driving opioid addiction. We found that glucocorticoid receptor (GR) antagonism with mifepristone reduced opioid addiction-like behaviors in rats and zebrafish of both sexes and decreased the firing of corticotropin-releasing factor neurons in the rat amygdala (i.e., a marker of brain stress system activation). In support of the hypothesized role of glucocorticoid transcriptional regulation of extrahypothalamic GRs in addiction-like behavior, an intra-amygdala infusion of an antisense oligonucleotide that blocked GR transcriptional activity reduced addiction-like behaviors. Finally, we identified transcriptional adaptations of GR signaling in the amygdala of humans with OUD. Thus, GRs, their coregulators, and downstream systems may represent viable therapeutic targets to treat the "stress side" of OUD.


Subject(s)
Opioid-Related Disorders , Substance Withdrawal Syndrome , Adrenal Cortex Hormones , Animals , Corticotropin-Releasing Hormone , Rats , Zebrafish
3.
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
4.
Mol Ther ; 28(12): 2662-2676, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32818431

ABSTRACT

Usher syndrome is a syndromic form of hereditary hearing impairment that includes sensorineural hearing loss and delayed-onset retinitis pigmentosa (RP). Type 1 Usher syndrome (USH1) is characterized by congenital profound sensorineural hearing impairment and vestibular areflexia, with adolescent-onset RP. Systemic treatment with antisense oligonucleotides (ASOs) targeting the human USH1C c.216G>A splicing mutation in a knockin mouse model of USH1 restores hearing and balance. Herein, we explore the effect of delivering ASOs locally to the ear to treat hearing and vestibular dysfunction associated with Usher syndrome. Three localized delivery strategies were investigated in USH1C mice: inner ear injection, trans-tympanic membrane injection, and topical tympanic membrane application. We demonstrate, for the first time, that ASOs delivered directly to the ear correct Ush1c expression in inner ear tissue, improve cochlear hair cell transduction currents, restore vestibular afferent irregularity, spontaneous firing rate, and sensitivity to head rotation, and successfully recover hearing thresholds and balance behaviors in USH1C mice. We conclude that local delivery of ASOs to the middle and inner ear reach hair cells and can rescue both hearing and balance. These results also demonstrate the therapeutic potential of ASOs to treat hearing and balance deficits associated with Usher syndrome and other ear diseases.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Ear, Middle/drug effects , Genetic Therapy/methods , Hair Cells, Auditory/drug effects , Mutation , Oligonucleotides, Antisense/administration & dosage , Usher Syndromes/genetics , Usher Syndromes/therapy , Vestibule, Labyrinth/drug effects , Administration, Topical , Animals , Animals, Newborn , Disease Models, Animal , Female , Gene Knock-In Techniques , Hair Cells, Auditory/metabolism , Hearing/drug effects , Injections , Male , Mice , Mice, Inbred C57BL , Tympanic Membrane/drug effects , Vestibule, Labyrinth/metabolism
5.
Hum Mol Genet ; 26(18): 3482-3494, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28633508

ABSTRACT

Usher syndrome type 1C (USH1C/harmonin) is associated with profound retinal, auditory and vestibular dysfunction. We have previously reported on an antisense oligonucleotide (ASO-29) that dramatically improves auditory function and balance behavior in mice homozygous for the harmonin mutation Ush1c c.216G > A following a single systemic administration. The findings were suggestive of improved vestibular function; however, no direct vestibular assessment was made. Here, we measured vestibular sensory evoked potentials (VsEPs) to directly assess vestibular function in Usher mice. We report that VsEPs are absent or abnormal in Usher mice, indicating profound loss of vestibular function. Strikingly, Usher mice receiving ASO-29 treatment have normal or elevated vestibular response thresholds when treated during a critical period between postnatal day 1 and 5, respectively. In contrast, treatment of mice with ASO-29 treatment at P15 was minimally effective at rescuing vestibular function. Interestingly, ASO-29 treatment at P1, P5 or P15 resulted in sufficient vestibular recovery to support normal balance behaviors, suggesting a therapeutic benefit to balance with ASO-29 treatment at P15 despite the profound vestibular functional deficits that persist with treatment at this later time. These findings provide the first direct evidence of an effective treatment of peripheral vestibular function in a mouse model of USH1C and reveal the potential for using antisense technology to treat vestibular dysfunction.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Usher Syndromes/therapy , Animals , Cell Cycle Proteins , Cytoskeletal Proteins , Disease Models, Animal , Evoked Potentials, Auditory , Hearing/genetics , Mice , Mutation , Oligonucleotides, Antisense/therapeutic use , Retina/metabolism , Retinal Degeneration/genetics , Usher Syndromes/genetics , Usher Syndromes/metabolism , Vestibular Evoked Myogenic Potentials/genetics , Vestibule, Labyrinth/metabolism , Vestibule, Labyrinth/physiology
6.
Mol Cell ; 43(6): 927-39, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925381

ABSTRACT

Alternative intronic polyadenylation (IPA) can generate truncated protein isoforms with significantly altered functions. Here, we describe 31 dominant-negative, secreted variant isoforms of receptor tyrosine kinases (RTKs) that are produced by activation of intronic poly(A) sites. We show that blocking U1-snRNP can activate IPA, indicating a larger role for U1-snRNP in RNA surveillance. Moreover, we report the development of an antisense-based method to effectively and specifically activate expression of individual soluble decoy RTKs (sdRTKs) to alter signaling, with potential therapeutic implications. In particular, a quantitative switch from signal transducing full-length vascular endothelial growth factor receptor-2 (VEGFR2/KDR) to a dominant-negative sKDR results in a strong antiangiogenic effect both on directly targeted cells and on naive cells exposed to conditioned media, suggesting a role for this approach in interfering with angiogenic paracrine and autocrine loops.


Subject(s)
Introns , Polyadenylation , Receptor Protein-Tyrosine Kinases/biosynthesis , Humans , Neovascularization, Physiologic/physiology , Poly A/chemistry , Poly A/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/physiology , RNA Splicing , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/physiology
7.
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
8.
Am J Hum Genet ; 93(5): 798-811, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24140112

ABSTRACT

Copy-number variants (CNVs) represent a significant interpretative challenge, given that each CNV typically affects the dosage of multiple genes. Here we report on five individuals with coloboma, microcephaly, developmental delay, short stature, and craniofacial, cardiac, and renal defects who harbor overlapping microdeletions on 8q24.3. Fine mapping localized a commonly deleted 78 kb region that contains three genes: SCRIB, NRBP2, and PUF60. In vivo dissection of the CNV showed discrete contributions of the planar cell polarity effector SCRIB and the splicing factor PUF60 to the syndromic phenotype, and the combinatorial suppression of both genes exacerbated some, but not all, phenotypic components. Consistent with these findings, we identified an individual with microcephaly, short stature, intellectual disability, and heart defects with a de novo c.505C>T variant leading to a p.His169Tyr change in PUF60. Functional testing of this allele in vivo and in vitro showed that the mutation perturbs the relative dosage of two PUF60 isoforms and, subsequently, the splicing efficiency of downstream PUF60 targets. These data inform the functions of two genes not associated previously with human genetic disease and demonstrate how CNVs can exhibit complex genetic architecture, with the phenotype being the amalgam of both discrete dosage dysfunction of single transcripts and also of binary genetic interactions.


Subject(s)
DNA Copy Number Variations , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Alleles , Animals , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , Developmental Disabilities/genetics , Female , Gene Deletion , Gene Knockdown Techniques , HeLa Cells , Humans , Intellectual Disability/genetics , Male , Microcephaly/genetics , Phenotype , RNA Splicing Factors , Zebrafish/genetics
9.
Neuroscience ; 511: 53-69, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36587866

ABSTRACT

Alzheimer's disease (AD) is associated with hippocampal neuropathology and cognitive impairments, including wandering behavior or becoming lost in a familiar environment. Wandering behavior is severe and manifests early in life for people with specific genetic mutations. Genetic mouse models of AD have been developed to characterize the onset and progression of behavioral deficits that represent human behaviors, such as wandering, to test the efficacy of therapeutics. It is not clear if current assessments of mouse models capture the onset of AD or a snapshot of its progression. Sequential analysis of open field behavior provides a robust, quick test to dissociate navigation cues that contribute to spatial disorientation, a feature of wandering. Despite potential utility in evaluating this feature of AD, little work has been reported using animal models of dementia in this task. Thus, we examined the use of different sources of information to maintain spatial orientation at two prodromal ages in female transgenic CRND8 AD (n = 17) and Control mice (n = 16). These mice exhibit amyloid plaques, a hallmark neuropathological feature of AD, that are associated with cognitive dysfunction at ∼three months of age. Spatial disorientation was observed at two months and more severely at four months under dark conditions, but performance was spared when visual environmental cues were available. This study provides documentation of impaired self-movement cue processing in AD mice, establishing the dark open field as a behavioral tool to characterize spatial disorientation associated with AD. These findings may accelerate future assessments of novel therapeutic interventions for neurological disorders.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Mice , Animals , Alzheimer Disease/pathology , Confusion , Hippocampus/metabolism , Disease Models, Animal , Mice, Transgenic , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism
10.
Hum Mol Genet ; 19(24): 4906-17, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20884664

ABSTRACT

Spinal muscular atrophy (SMA) is a neurological disorder characterized by motor neuron degeneration and progressive muscle paralysis. The disease is caused by a reduction in survival of motor neuron (SMN) protein resulting from homozygous deletion of the SMN1 gene. SMN protein is also encoded by SMN2. However, splicing of SMN2 exon 7 is defective, and consequently, the majority of the transcripts produce a truncated, unstable protein. SMN protein itself has a role in splicing. The protein is required for the biogenesis of spliceosomal snRNPs, which are essential components of the splicing reaction. We now show that SMN protein abundance affects the splicing of SMN2 exon 7, revealing a feedback loop inSMN expression. The reduced SMN protein concentration observed in SMA samples and in cells depleted of SMN correlates with a decrease in cellular snRNA levels and a decrease in SMN2 exon 7 splicing. Furthermore, altering the relative abundance or activity of individual snRNPs has distinct effects on exon 7 splicing, demonstrating that core spliceosomal snRNPs influence SMN2 alternative splicing. Our results identify a feedback loop in SMN expression by which low SMN protein levels exacerbate SMN exon 7 skipping, leading to a further reduction in SMN protein. These results imply that a modest increase in SMN protein abundance may cause a disproportionately large increase in SMN expression, a finding that is important for assessing the therapeutic potential of SMA treatments and understanding disease pathogenesis.


Subject(s)
Feedback, Physiological , Muscular Atrophy, Spinal/genetics , RNA Splicing/genetics , Animals , Binding Sites , Exons/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U1 Small Nuclear/genetics , Survival of Motor Neuron 2 Protein/genetics
11.
Nat Med ; 26(9): 1444-1451, 2020 09.
Article in English | MEDLINE | ID: mdl-32719489

ABSTRACT

CLN3 Batten disease is an autosomal recessive, neurodegenerative, lysosomal storage disease caused by mutations in CLN3, which encodes a lysosomal membrane protein1-3. There are no disease-modifying treatments for this disease that affects up to 1 in 25,000 births, has an onset of symptoms in early childhood and typically is fatal by 20-30 years of life4-7. Most patients with CLN3 Batten have a deletion encompassing exons 7 and 8 (CLN3∆ex7/8), creating a reading frameshift7,8. Here we demonstrate that mice with this deletion can be effectively treated using an antisense oligonucleotide (ASO) that induces exon skipping to restore the open reading frame. A single treatment of neonatal mice with an exon 5-targeted ASO-induced robust exon skipping for more than a year, improved motor coordination, reduced histopathology in Cln3∆ex7/8 mice and increased survival in a new mouse model of the disease. ASOs also induced exon skipping in cell lines derived from patients with CLN3 Batten disease. Our findings demonstrate the utility of ASO-based reading-frame correction as an approach to treat CLN3 Batten disease and broaden the therapeutic landscape for ASOs in the treatment of other diseases using a similar strategy.


Subject(s)
Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/genetics , Oligonucleotides, Antisense/therapeutic use , Animals , Cell Line , Codon, Nonsense/genetics , Disease Models, Animal , Frameshift Mutation/genetics , Humans , Mice
12.
Stem Cell Reports ; 12(1): 29-41, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30595548

ABSTRACT

The Parkinson disease (PD) genetic LRRK2 gain-of-function mutations may relate to the ER pathological changes seen in PD patients at postmortem. Human induced pluripotent stem cell (iPSC)-derived neurons with the PD pathogenic LRRK2 G2019S mutation exhibited neurite collapse when challenged with the ER Ca2+ influx sarco/ER Ca2+-ATPase inhibitor thapsigargin (THP). Baseline ER Ca2+ levels measured with the ER Ca2+ indicator CEPIA-ER were lower in LRRK2 G2019S human neurons, including in differentiated midbrain dopamine neurons in vitro. After THP challenge, PD patient-derived neurons displayed increased Ca2+ influx and decreased intracellular Ca2+ buffering upon membrane depolarization. These effects were reversed following LRRK2 mutation correction by antisense oligonucleotides and gene editing. Gene expression analysis in LRRK2 G2019S neurons identified modified levels of key store-operated Ca2+ entry regulators, with no alterations in ER Ca2+ efflux. These results demonstrate PD gene mutation LRRK2 G2019S ER calcium-dependent pathogenic effects in human neurons.


Subject(s)
Calcium Signaling , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Neurites/metabolism , Parkinson Disease/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Humans , Mutation, Missense , Neurites/drug effects , Neurites/pathology , Parkinson Disease/genetics , Thapsigargin/pharmacology
13.
Cancer Res ; 66(10): 5181-9, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707442

ABSTRACT

The BRCA1 tumor suppressor contributes to the repair of DNA double-strand breaks (DSB) through homologous recombination, but the mechanism is unknown. The rapid accumulation of BRCA1 into nuclear foci in response to induction of DNA breaks suggests that BRCA1 may function in an early step in the repair pathway. We examined the role of BRCA1 in one such early step, the resection of DSBs to generate ssDNA. The appearance of ssDNA in response to ionizing radiation is similar to that of BRCA1 foci formation, suggesting that the two processes are related. Furthermore, BRCA1 colocalizes to ssDNA sites induced by ionizing radiation. Overexpression of BRCA1 causes an increase in cells exhibiting ssDNA induced by ionizing radiation. Mutant BRCA1 that lacks the COOH-terminal BRCT domain also promotes ssDNA but fails to form nuclear foci. Knockdown of BRCA1 expression reduces ssDNA and Rad51 foci formation in response to ionizing radiation. These results indicate that BRCA1 promotes induction of ssDNA in response to ionizing radiation and accumulates at sites of ssDNA.


Subject(s)
BRCA1 Protein/metabolism , DNA, Single-Stranded/biosynthesis , DNA, Single-Stranded/radiation effects , BRCA1 Protein/biosynthesis , BRCA1 Protein/genetics , Breast/cytology , Breast/metabolism , Breast/radiation effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Repair , Humans
14.
J Assoc Res Otolaryngol ; 19(1): 1-16, 2018 02.
Article in English | MEDLINE | ID: mdl-29027038

ABSTRACT

The absence of functional outer hair cells is a component of several forms of hereditary hearing impairment, including Usher syndrome, the most common cause of concurrent hearing and vision loss. Antisense oligonucleotide (ASO) treatment of mice with the human Usher mutation, Ush1c c.216G>A, corrects gene expression and significantly improves hearing, as measured by auditory-evoked brainstem responses (ABRs), as well as inner and outer hair cell (IHC and OHC) bundle morphology. However, it is not clear whether the improvement in hearing achieved by ASO treatment involves the functional rescue of outer hair cells. Here, we show that Ush1c c.216AA mice lack OHC function as evidenced by the absence of distortion product otoacoustic emissions (DPOAEs) in response to low-, mid-, and high-frequency tone pairs. This OHC deficit is rescued by treatment with an ASO that corrects expression of Ush1c c.216G>A. Interestingly, although rescue of inner hairs cells, as measured by ABR, is achieved by ASO treatment as late as 7 days after birth, rescue of outer hair cells, measured by DPOAE, requires treatment before post-natal day 5. These results suggest that ASO-mediated rescue of both IHC and OHC function is age dependent and that the treatment window is different for the different cell types. The timing of treatment for congenital hearing disorders is of critical importance for the development of drugs such ASO-29 for hearing rescue.


Subject(s)
Hair Cells, Auditory, Outer/drug effects , Oligonucleotides, Antisense/therapeutic use , Usher Syndromes/drug therapy , Age Factors , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cytoskeletal Proteins , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Outer/physiology , Mice , Mutation , Oligonucleotides, Antisense/pharmacology , Otoacoustic Emissions, Spontaneous , Usher Syndromes/physiopathology
15.
Behav Brain Res ; 338: 76-87, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29037661

ABSTRACT

Usher syndrome, Type 1C (USH1C) is an autosomal recessive inherited disorder in which a mutation in the gene encoding harmonin is associated with multi-sensory deficits (i.e., auditory, vestibular, and visual). USH1C (Usher) mice, engineered with a human USH1C mutation, exhibit these multi-sensory deficits by circling behavior and lack of response to sound. Administration of an antisense oligonucleotide (ASO) therapeutic that corrects expression of the mutated USH1C gene, has been shown to increase harmonin levels, reduce circling behavior, and improve vestibular and auditory function. The current study evaluates the organization of exploratory movements to assess spatial organization in Usher mice and determine the efficacy of ASO therapy in attenuating any such deficits. Usher and heterozygous mice received the therapeutic ASO, ASO-29, or a control, non-specific ASO treatment at postnatal day five. Organization of exploratory movements was assessed under dark and light conditions at two and six-months of age. Disruptions in exploratory movement organization observed in control-treated Usher mice were consistent with impaired use of self-movement and environmental cues. In general, ASO-29 treatment rescued organization of exploratory movements at two and six-month testing points. These observations are consistent with ASO-29 rescuing processing of multiple sources of information and demonstrate the potential of ASO therapies to ameliorate topographical disorientation associated with other genetic disorders.


Subject(s)
Carrier Proteins/genetics , Exploratory Behavior/drug effects , Movement/drug effects , Oligonucleotides, Antisense/pharmacology , Usher Syndromes/physiopathology , Animals , Behavior, Animal/drug effects , Carrier Proteins/metabolism , Cell Cycle Proteins , Cytoskeletal Proteins , Male , Mice , Usher Syndromes/genetics , Usher Syndromes/metabolism
16.
Brain Res ; 1182: 1-10, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-17936734

ABSTRACT

Most cases of Parkinson's disease (PD) are sporadic, suggesting an environmental influence on individuals affected by this neurodegenerative disorder. Environmental stresses often lead to changes in the regulation of splicing of pre-mRNA transcripts and this may lead to the pathogenesis of the disease. A 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid mouse model was used to examine the changes in the splicing of the fosB and rgs9 transcripts. The ratio of DeltafosB/fosB transcript was decreased in the substantia nigra and unchanged in the striatum after acute MPTP treatment. The DeltafosB/fosB transcript ratio decreased initially and then increased in the striatum of chronically MPTP-treated animals due to different degrees of reduction for the splice variants over time, whereas the ratio was unchanged in the substantia nigra. The ratio of rgs9-2/rgs9-1 transcript decreased in the substantia nigra of mice after acute MPTP treatment and increased temporarily in the striatum after chronic MPTP treatment. There was an increase in the DeltaFosB/FosB and RGS9-2/RGS9-1 protein ratios 3 weeks and 3 days post-treatment, respectively, in chronically treated mice. The data indicate that the pattern of splice isoforms of fosB and rgs9 reflects the brain's immediate and long-term responses to the physiological stress associated with Parkinsonism.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Dopamine Agents/administration & dosage , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RGS Proteins/metabolism , Animals , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Drug Administration Schedule , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fos/genetics , RGS Proteins/genetics , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
17.
Nat Biotechnol ; 35(3): 264-272, 2017 03.
Article in English | MEDLINE | ID: mdl-28165476

ABSTRACT

Because there are currently no biological treatments for hearing loss, we sought to advance gene therapy approaches to treat genetic deafness. We focused on Usher syndrome, a devastating genetic disorder that causes blindness, balance disorders and profound deafness, and studied a knock-in mouse model, Ush1c c.216G>A, for Usher syndrome type IC (USH1C). As restoration of complex auditory and balance function is likely to require gene delivery systems that target auditory and vestibular sensory cells with high efficiency, we delivered wild-type Ush1c into the inner ear of Ush1c c.216G>A mice using a synthetic adeno-associated viral vector, Anc80L65, shown to transduce 80-90% of sensory hair cells. We demonstrate recovery of gene and protein expression, restoration of sensory cell function, rescue of complex auditory function and recovery of hearing and balance behavior to near wild-type levels. The data represent unprecedented recovery of inner ear function and suggest that biological therapies to treat deafness may be suitable for translation to humans with genetic inner ear disorders.


Subject(s)
Carrier Proteins/genetics , Genetic Therapy/methods , Hearing Loss, Sensorineural/therapy , Usher Syndromes/genetics , Usher Syndromes/therapy , Vestibular Diseases/therapy , Animals , Cell Cycle Proteins , Cytoskeletal Proteins , Female , Gene Knock-In Techniques , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Male , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/genetics , Recovery of Function/genetics , Treatment Outcome , Vestibular Diseases/diagnosis , Vestibular Diseases/genetics
18.
EMBO Mol Med ; 8(4): 328-45, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26902204

ABSTRACT

Apolipoprotein E receptor 2 (ApoER2) is an apolipoprotein E receptor involved in long-term potentiation, learning, and memory. Given its role in cognition and its association with the Alzheimer's disease (AD) risk gene, apoE, ApoER2 has been proposed to be involved in AD, though a role for the receptor in the disease is not clear. ApoER2 signaling requires amino acids encoded by alternatively spliced exon 19. Here, we report that the balance of ApoER2 exon 19 splicing is deregulated in postmortem brain tissue from AD patients and in a transgenic mouse model of AD To test the role of deregulated ApoER2 splicing in AD, we designed an antisense oligonucleotide (ASO) that increases exon 19 splicing. Treatment of AD mice with a single dose of ASO corrected ApoER2 splicing for up to 6 months and improved synaptic function and learning and memory. These results reveal an association between ApoER2 isoform expression and AD, and provide preclinical evidence for the utility of ASOs as a therapeutic approach to mitigate Alzheimer's disease symptoms by improving ApoER2 exon 19 splicing.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , LDL-Receptor Related Proteins/metabolism , Oligonucleotides, Antisense/therapeutic use , RNA Splicing , Alzheimer Disease/pathology , Animals , Brain/physiology , Disease Models, Animal , Humans , LDL-Receptor Related Proteins/genetics , Learning , Memory , Mice , Mice, Transgenic , Oligonucleotides, Antisense/genetics , Treatment Outcome
19.
PLoS One ; 9(12): e115205, 2014.
Article in English | MEDLINE | ID: mdl-25506695

ABSTRACT

Spinal muscular atrophy (SMA) is one of the most common inherited causes of pediatric mortality. SMA is caused by deletions or mutations in the survival of motor neuron 1 (SMN1) gene, which results in SMN protein deficiency. Humans have a centromeric copy of the survival of motor neuron gene, SMN2, which is nearly identical to SMN1. However, SMN2 cannot compensate for the loss of SMN1 because SMN2 has a single-nucleotide difference in exon 7, which negatively affects splicing of the exon. As a result, most mRNA produced from SMN2 lacks exon 7. SMN2 mRNA lacking exon 7 encodes a truncated protein with reduced functionality. Improving SMN2 exon 7 inclusion is a goal of many SMA therapeutic strategies. The identification of regulators of exon 7 inclusion may provide additional therapeutic targets or improve the design of existing strategies. Although a number of regulators of exon 7 inclusion have been identified, the function of most splicing proteins in exon 7 inclusion is unknown. Here, we test the role of SR proteins and hnRNP proteins in SMN2 exon 7 inclusion. Knockdown and overexpression studies reveal that SRSF1, SRSF2, SRSF3, SRSF4, SRSF5, SRSF6, SRSF7, SRSF11, hnRNPA1/B1 and hnRNP U can inhibit exon 7 inclusion. Depletion of two of the most potent inhibitors of exon 7 inclusion, SRSF2 or SRSF3, in cell lines derived from SMA patients, increased SMN2 exon 7 inclusion and SMN protein. Our results identify novel regulators of SMN2 exon 7 inclusion, revealing potential targets for SMA therapeutics.


Subject(s)
Muscular Atrophy, Spinal/genetics , Nuclear Proteins/physiology , RNA-Binding Proteins/physiology , Ribonucleoproteins/physiology , Cell Line , Exons , Female , Gene Expression , Gene Knockdown Techniques , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Humans , Muscular Atrophy, Spinal/physiopathology , Nuclear Proteins/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/physiology , Survival of Motor Neuron 2 Protein/genetics
20.
Nat Med ; 19(3): 345-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23380860

ABSTRACT

Hearing impairment is the most common sensory disorder, with congenital hearing impairment present in approximately 1 in 1,000 newborns. Hereditary deafness is often mediated by the improper development or degeneration of cochlear hair cells. Until now, it was not known whether such congenital failures could be mitigated by therapeutic intervention. Here we show that hearing and vestibular function can be rescued in a mouse model of human hereditary deafness. An antisense oligonucleotide (ASO) was used to correct defective pre-mRNA splicing of transcripts from the USH1C gene with the c.216G>A mutation, which causes human Usher syndrome, the leading genetic cause of combined deafness and blindness. Treatment of neonatal mice with a single systemic dose of ASO partially corrects Ush1c c.216G>A splicing, increases protein expression, improves stereocilia organization in the cochlea, and rescues cochlear hair cells, vestibular function and low-frequency hearing in mice. These effects were sustained for several months, providing evidence that congenital deafness can be effectively overcome by treatment early in development to correct gene expression and demonstrating the therapeutic potential of ASOs in the treatment of deafness.


Subject(s)
Carrier Proteins/genetics , Oligonucleotides, Antisense/therapeutic use , Usher Syndromes/genetics , Usher Syndromes/therapy , Animals , Cell Cycle Proteins , Cell Line, Tumor , Cochlea/metabolism , Cytoskeletal Proteins , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Genetic Therapy , Hair Cells, Auditory/metabolism , HeLa Cells , Hearing , Humans , Mice , Stereocilia/genetics , Usher Syndromes/metabolism , Vestibule, Labyrinth/physiology
SELECTION OF CITATIONS
SEARCH DETAIL