Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
Clin Obstet Gynecol ; 66(3): 616-623, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37650671

ABSTRACT

The utilization of cell-free DNA (cfDNA) screening has expanded rapidly across the age spectrum of pregnant persons. With cfDNA's widespread adoption, genetic fetal sex is now often known before a phenotypic assessment on anatomic survey. CfDNA detects sex discordance in 1/1500 to 2000 pregnancies. Upon detection of sex discordance, lab error or other factors should first be assessed. Once other causes have been ruled out, this may indicate an underlying disorder/difference in sex development. A multidisciplinary team should coordinate diagnosis, treatment, and support for the family. This review discusses the diagnostic workup, emphasizing the multidisciplinary counseling and management of disorder/differences in sex development.


Subject(s)
Cell-Free Nucleic Acids , Prenatal Care , Female , Pregnancy , Humans , Sexual Development
3.
Am J Obstet Gynecol MFM ; 5(7): 100997, 2023 07.
Article in English | MEDLINE | ID: mdl-37271593

ABSTRACT

BACKGROUND: Nausea and vomiting is one of the most common complications of pregnancy, affecting 50% to 80% of pregnant persons. Moreover, despite its prevalence, it remains a challenging condition to treat. Treatment often involves oral and intravenous medications with potential side effects, particularly when taken in combination. Capsaicin cream is proven to decrease nausea and vomiting in cyclic vomiting syndrome; however, its use has not been well studied among pregnant patients. OBJECTIVE: This study aimed to test the feasibility of the off-label use of capsaicin for the treatment of nausea and vomiting in pregnancy. STUDY DESIGN: This was a double-blinded randomized controlled trial of pregnant individuals in their first trimester of pregnancy seeking care at a tertiary care hospital for nausea and vomiting. Consenting participants were randomized to abdominal application of topical capsaicin vs placebo. All participants received intravenous hydration and metoclopramide. The primary outcome, total treatment time, was recorded for all participants. In addition, symptom severity was assessed every 30 minutes using a visual analog scale. Data were analyzed using the Wilcoxon rank-sum test for continuous variables and the Fisher exact test for binary variables. RESULTS: Of the 38 eligible individuals approached, 30 were randomized. There was a trend toward decreased mean treatment time in the capsaicin group compared with the placebo group (79.9 vs 97.3 minutes; P=.1). There was no significant difference in visual analog scale scores at any time point between groups. Furthermore, capsaicin was well tolerated, with only 1 individual requesting the medication be removed. CONCLUSION: This study demonstrated that capsaicin is an acceptable treatment of nausea and vomiting in pregnancy and additional explorations of its use as treatment are feasible. A larger randomized controlled trial is needed to determine the efficacy of capsaicin in this population.


Subject(s)
Antiemetics , Pregnancy , Female , Humans , Antiemetics/adverse effects , Capsaicin/adverse effects , Pilot Projects , Vomiting/chemically induced , Vomiting/drug therapy , Vomiting/prevention & control , Nausea/chemically induced , Nausea/drug therapy , Nausea/prevention & control
4.
Breastfeed Med ; 16(8): 614-623, 2021 08.
Article in English | MEDLINE | ID: mdl-33826418

ABSTRACT

Background: Low-income women are less likely to breastfeed than high-income women. Technology-based interventions demonstrate promise in decreasing health disparities. We assessed whether increased use of breastfeeding smartphone applications (apps) impacts breastfeeding rates for low-income women. Materials and Methods: This is a secondary analysis of a randomized control trial (RCT), including nulliparous, low-income women. Women were randomized to one of two novel apps: control app containing digital breastfeeding handouts and BreastFeeding Friend (BFF), an interactive app containing on-demand breastfeeding educational and video content. App usage was securely tracked. The highest quartile of BFF and control app users were combined and compared to the lowest quartile of app users. The primary outcome was breastfeeding initiation. Secondary outcomes included breastfeeding outcomes and resource preferences through 6 months. Results: In the RCT, BFF and control app median uses were 15 (interquartile range [IQR] 4-24) and 9 (IQR 5-19) (p = 0.1), respectively. Breastfeeding initiation did not differ with app usage (84.1% in highest quartile versus 78.2% for lowest quartile; p = 0.5). Rates of sustained and exclusive breastfeeding through 6 months were similar between groups. Among both groups, smartphone apps were the most preferred breastfeeding resource at 6 weeks. Low quartile users also preferred alternative online breastfeeding resources: >50% of all users preferred technology-based breastfeeding resources. Conclusions: Increased usage of breastfeeding apps did not improve breastfeeding rates among low-income women. However, technology-based resources were the most preferred breastfeeding resource after hospital discharge, indicating ongoing development of technology-based interventions has potential to increase breastfeeding in this high-needs population. clinicaltrials.gov (NCT03167073).


Subject(s)
Breast Feeding , Mobile Applications , Cognition , Female , Humans , Smartphone
5.
J Clin Invest ; 129(12): 5568-5583, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31557132

ABSTRACT

Gene therapy approaches are being deployed to treat recessive genetic disorders by restoring the expression of mutated genes. However, the feasibility of these approaches for dominantly inherited diseases - where treatment may require reduction in the expression of a toxic mutant protein resulting from a gain-of-function allele - is unclear. Here we show the efficacy of allele-specific RNAi as a potential therapy for Charcot-Marie-Tooth disease type 2D (CMT2D), caused by dominant mutations in glycyl-tRNA synthetase (GARS). A de novo mutation in GARS was identified in a patient with a severe peripheral neuropathy, and a mouse model precisely recreating the mutation was produced. These mice developed a neuropathy by 3-4 weeks of age, validating the pathogenicity of the mutation. RNAi sequences targeting mutant GARS mRNA, but not wild-type, were optimized and then packaged into AAV9 for in vivo delivery. This almost completely prevented the neuropathy in mice treated at birth. Delaying treatment until after disease onset showed modest benefit, though this effect decreased the longer treatment was delayed. These outcomes were reproduced in a second mouse model of CMT2D using a vector specifically targeting that allele. The effects were dose dependent, and persisted for at least 1 year. Our findings demonstrate the feasibility of AAV9-mediated allele-specific knockdown and provide proof of concept for gene therapy approaches for dominant neuromuscular diseases.


Subject(s)
Charcot-Marie-Tooth Disease/therapy , Genetic Therapy , Glycine-tRNA Ligase/genetics , RNA Interference , Alleles , Animals , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mutation
6.
Hum Mol Genet ; 28(4): 525-538, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30304524

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are critical for protein translation. Pathogenic variants of ARSs have been previously associated with peripheral neuropathy and multisystem disease in heterozygotes and homozygotes, respectively. We report seven related children homozygous for a novel mutation in tyrosyl-tRNA synthetase (YARS, c.499C > A, p.Pro167Thr) identified by whole exome sequencing. This variant lies within a highly conserved interface required for protein homodimerization, an essential step in YARS catalytic function. Affected children expressed a more severe phenotype than previously reported, including poor growth, developmental delay, brain dysmyelination, sensorineural hearing loss, nystagmus, progressive cholestatic liver disease, pancreatic insufficiency, hypoglycemia, anemia, intermittent proteinuria, recurrent bloodstream infections and chronic pulmonary disease. Related adults heterozygous for YARS p.Pro167Thr showed no evidence of peripheral neuropathy on electromyography, in contrast to previous reports for other YARS variants. Analysis of YARS p.Pro167Thr in yeast complementation assays revealed a loss-of-function, hypomorphic allele that significantly impaired growth. Recombinant YARS p.Pro167Thr demonstrated normal subcellular localization, but greatly diminished ability to homodimerize in human embryonic kidney cells. This work adds to a rapidly growing body of research emphasizing the importance of ARSs in multisystem disease and significantly expands the allelic and clinical heterogeneity of YARS-associated human disease. A deeper understanding of the role of YARS in human disease may inspire innovative therapies and improve care of affected patients.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Loss of Function Mutation/genetics , Tyrosine-tRNA Ligase/genetics , Adult , Catalytic Domain/genetics , Child, Preschool , Female , Genetic Diseases, Inborn/physiopathology , Hearing Loss, Sensorineural/diagnostic imaging , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Mutation , Pedigree , Phenotype , Severity of Illness Index , Exome Sequencing , Yeasts/genetics
7.
Hum Mutat ; 39(6): 834-840, 2018 06.
Article in English | MEDLINE | ID: mdl-29573043

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed enzymes that ligate amino acids onto tRNA molecules. Genes encoding ARSs have been implicated in phenotypically diverse dominant and recessive human diseases. The charging of tRNAPHE with phenylalanine is performed by a tetrameric enzyme that contains two alpha (FARSA) and two beta (FARSB) subunits. To date, mutations in the genes encoding these subunits (FARSA and FARSB) have not been implicated in any human disease. Here, we describe a patient with a severe, lethal, multisystem, developmental phenotype who was compound heterozygous for FARSB variants: p.Thr256Met and p.His496Lysfs*14. Expression studies using fibroblasts isolated from the proband revealed a severe depletion of both FARSB and FARSA protein levels. These data indicate that the FARSB variants destabilize total phenylalanyl-tRNA synthetase levels, thus causing a loss-of-function effect. Importantly, our patient shows strong phenotypic overlap with patients that have recessive diseases associated with other ARS loci; these observations strongly support the pathogenicity of the identified FARSB variants and are consistent with the essential function of phenylalanyl-tRNA synthetase in human cells. In sum, our clinical, genetic, and functional analyses revealed the first FARSB variants associated with a human disease phenotype and expand the locus heterogeneity of ARS-related human disease.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Genetic Predisposition to Disease , Loss of Function Mutation/genetics , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/deficiency , Gene Expression Regulation , Humans , Male , Phenotype , Phenylalanine-tRNA Ligase/genetics
8.
Hum Mutat ; 39(3): 415-432, 2018 03.
Article in English | MEDLINE | ID: mdl-29235198

ABSTRACT

Histidyl-tRNA synthetase (HARS) ligates histidine to cognate tRNA molecules, which is required for protein translation. Mutations in HARS cause the dominant axonal peripheral neuropathy Charcot-Marie-Tooth disease type 2W (CMT2W); however, the precise molecular mechanism remains undefined. Here, we investigated three HARS missense mutations associated with CMT2W (p.Tyr330Cys, p.Ser356Asn, and p.Val155Gly). The three mutations localize to the HARS catalytic domain and failed to complement deletion of the yeast ortholog (HTS1). Enzyme kinetics, differential scanning fluorimetry (DSF), and analytical ultracentrifugation (AUC) were employed to assess the effect of these substitutions on primary aminoacylation function and overall dimeric structure. Notably, the p.Tyr330Cys, p.Ser356Asn, and p.Val155Gly HARS substitutions all led to reduced aminoacylation, providing a direct connection between CMT2W-linked HARS mutations and loss of canonical ARS function. While DSF assays revealed that only one of the variants (p.Val155Gly) was less thermally stable relative to wild-type, all three HARS mutants formed stable dimers, as measured by AUC. Our work represents the first biochemical analysis of CMT-associated HARS mutations and underscores how loss of the primary aminoacylation function can contribute to disease pathology.


Subject(s)
Axons/pathology , Histidine-tRNA Ligase/metabolism , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/pathology , Amino Acid Sequence , Aminoacylation , Biocatalysis , Catalytic Domain , Conserved Sequence , Female , Genetic Complementation Test , Histidine-tRNA Ligase/chemistry , Histidine-tRNA Ligase/genetics , Histidine-tRNA Ligase/isolation & purification , Humans , Kinetics , Male , Mutation/genetics , Pedigree , Peripheral Nervous System Diseases/genetics , Protein Multimerization , Substrate Specificity
9.
Methods ; 113: 139-151, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27876679

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed, essential enzymes responsible for charging tRNA with cognate amino acids-the first step in protein synthesis. ARSs are required for protein translation in the cytoplasm and mitochondria of all cells. Surprisingly, mutations in 28 of the 37 nuclear-encoded human ARS genes have been linked to a variety of recessive and dominant tissue-specific disorders. Current data indicate that impaired enzyme function is a robust predictor of the pathogenicity of ARS mutations. However, experimental model systems that distinguish between pathogenic and non-pathogenic ARS variants are required for implicating newly identified ARS mutations in disease. Here, we outline strategies to assist in predicting the pathogenicity of ARS variants and urge cautious evaluation of genetic and functional data prior to linking an ARS mutation to a human disease phenotype.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Genetic Predisposition to Disease , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Mutation , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cytoplasm/genetics , Cytoplasm/metabolism , Disease Models, Animal , Gene Expression , Genetic Linkage , Hereditary Sensory and Motor Neuropathy/enzymology , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Pedigree , Penetrance , Phenotype , Prognosis
10.
Hum Mol Genet ; 25(8): 1528-42, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27008886

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a genetically heterogeneous group of peripheral neuropathies. Mutations in several aminoacyl-tRNA synthetase (ARS) genes have been implicated in inherited CMT disease. There are 12 reported CMT-causing mutations dispersed throughout the primary sequence of the human glycyl-tRNA synthetase (GARS). While there is strong genetic evidence linking GARS mutations to CMT disease, the molecular pathology underlying the neuromuscular and sensory phenotypes is still not fully understood. In particular, it is unclear whether the mutations result in a toxic gain of function, a partial loss of activity related to translation, or a combination of these mechanisms. We identified a zebrafish allele of gars (gars(s266)). Homozygous mutant embryos carry a C->A transversion, that changes a threonine to a lysine, in a residue next to a CMT-associated human mutation. We show that the neuromuscular phenotype observed in animals homozygous for T209K Gars (T130K in GARS) is due to a loss of dimerization of the mutated protein. Furthermore, we show that the loss of function, dimer-deficient and human disease-associated G319R Gars (G240R in GARS) mutant protein is unable to rescue the above phenotype. Finally, we demonstrate that another human disease-associated mutant G605R Gars (G526 in GARS) dimerizes with the remaining wild-type protein in animals heterozygous for the T209K Gars and reduces the function enough to elicit a neuromuscular phenotype. Our data indicate that dimerization is required for the dominant neurotoxicity of disease-associated GARS mutations and provide a rapid, tractable model for studying newly identified GARS variants for a role in human disease.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/genetics , Mutation , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Animals , Cells, Cultured , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Gene Expression Regulation , Glycine-tRNA Ligase/metabolism , Humans , Models, Biological , Phenotype , Protein Multimerization , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism
11.
Cell Rep ; 12(7): 1169-83, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26257172

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous distal symmetric polyneuropathy. Whole-exome sequencing (WES) of 40 individuals from 37 unrelated families with CMT-like peripheral neuropathy refractory to molecular diagnosis identified apparent causal mutations in ∼ 45% (17/37) of families. Three candidate disease genes are proposed, supported by a combination of genetic and in vivo studies. Aggregate analysis of mutation data revealed a significantly increased number of rare variants across 58 neuropathy-associated genes in subjects versus controls, confirmed in a second ethnically discrete neuropathy cohort, suggesting that mutation burden potentially contributes to phenotypic variability. Neuropathy genes shown to have highly penetrant Mendelizing variants (HPMVs) and implicated by burden in families were shown to interact genetically in a zebrafish assay exacerbating the phenotype established by the suppression of single genes. Our findings suggest that the combinatorial effect of rare variants contributes to disease burden and variable expressivity.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Exome , Genetic Load , Peripheral Nervous System Diseases/genetics , Phenotype , Animals , Female , Genetic Variation , HSP40 Heat-Shock Proteins/genetics , Humans , Male , Mutation , Myelin P2 Protein/genetics , Pedigree , Penetrance , Serine C-Palmitoyltransferase/genetics , Suppression, Genetic , Zebrafish
12.
Nat Genet ; 47(8): 926-32, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168012

ABSTRACT

Dominant optic atrophy (DOA) and axonal peripheral neuropathy (Charcot-Marie-Tooth type 2, or CMT2) are hereditary neurodegenerative disorders most commonly caused by mutations in the canonical mitochondrial fusion genes OPA1 and MFN2, respectively. In yeast, homologs of OPA1 (Mgm1) and MFN2 (Fzo1) work in concert with Ugo1, for which no human equivalent has been identified thus far. By whole-exome sequencing of patients with optic atrophy and CMT2, we identified four families with recessive mutations in SLC25A46. We demonstrate that SLC25A46, like Ugo1, is a modified carrier protein that has been recruited to the outer mitochondrial membrane and interacts with the inner membrane remodeling protein mitofilin (Fcj1). Loss of function in cultured cells and in zebrafish unexpectedly leads to increased mitochondrial connectivity, while severely affecting the development and maintenance of neurons in the fish. The discovery of SLC25A46 strengthens the genetic overlap between optic atrophy and CMT2 while exemplifying a new class of modified solute transporters linked to mitochondrial dynamics.


Subject(s)
Genetic Predisposition to Disease/genetics , Mitochondrial Proteins/genetics , Mutation , Optic Atrophy, Autosomal Dominant/genetics , Phosphate Transport Proteins/genetics , Animals , Animals, Genetically Modified , COS Cells , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Chlorocebus aethiops , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Exome/genetics , Female , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Confocal , Microscopy, Electron, Transmission , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Optic Atrophy, Autosomal Dominant/metabolism , Optic Atrophy, Autosomal Dominant/pathology , Pedigree , Phosphate Transport Proteins/metabolism , Protein Binding , RNA Interference , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Analysis, DNA , Zebrafish/embryology , Zebrafish/metabolism
13.
Neurology ; 84(20): 2040-7, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25904691

ABSTRACT

OBJECTIVE: To determine the genetic cause of neurodegeneration in a family with myeloneuropathy. METHODS: We studied 5 siblings in a family with a mild, dominantly inherited neuropathy by clinical examination and electrophysiology. One patient had a sural nerve biopsy. After ruling out common genetic causes of axonal Charcot-Marie-Tooth disease, we sequenced 3 tRNA synthetase genes associated with neuropathy. RESULTS: All affected family members had a mild axonal neuropathy, and 3 of 4 had lower extremity hyperreflexia, evidence of a superimposed myelopathy. A nerve biopsy showed evidence of chronic axonal loss. All affected family members had a heterozygous missense mutation c.304G>C (p.Gly102Arg) in the alanyl-tRNA synthetase (AARS) gene; this allele was not identified in unaffected individuals or control samples. The equivalent change in the yeast ortholog failed to complement a strain of yeast lacking AARS function, suggesting that the mutation is damaging. CONCLUSION: A novel mutation in AARS causes a mild myeloneuropathy, a novel phenotype for patients with mutations in one of the tRNA synthetase genes.


Subject(s)
Alanine-tRNA Ligase/genetics , Charcot-Marie-Tooth Disease/genetics , Mutation , Adult , Axons/ultrastructure , Family , Female , Genes, Dominant , Humans , Male , Middle Aged , Pedigree , Sural Nerve/ultrastructure , Young Adult
14.
Am J Hum Genet ; 96(4): 675-81, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25817015

ABSTRACT

Mutations in genes encoding aminoacyl-tRNA synthetases are known to cause leukodystrophies and genetic leukoencephalopathies-heritable disorders that result in white matter abnormalities in the central nervous system. Here we report three individuals (two siblings and an unrelated individual) with severe infantile epileptic encephalopathy, clubfoot, absent deep tendon reflexes, extrapyramidal symptoms, and persistently deficient myelination on MRI. Analysis by whole exome sequencing identified mutations in the nuclear-encoded alanyl-tRNA synthetase (AARS) in these two unrelated families: the two affected siblings are compound heterozygous for p.Lys81Thr and p.Arg751Gly AARS, and the single affected child is homozygous for p.Arg751Gly AARS. The two identified mutations were found to result in a significant reduction in function. Mutations in AARS were previously associated with an autosomal-dominant inherited form of axonal neuropathy, Charcot-Marie-Tooth disease type 2N (CMT2N). The autosomal-recessive AARS mutations identified in the individuals described here, however, cause a severe infantile epileptic encephalopathy with a central myelin defect and peripheral neuropathy, demonstrating that defects of alanyl-tRNA charging can result in a wide spectrum of disease manifestations.


Subject(s)
Abnormalities, Multiple/genetics , Alanine-tRNA Ligase/genetics , Epilepsy/genetics , Models, Molecular , Myelin Sheath/pathology , Peripheral Nervous System Diseases/genetics , Phenotype , Abnormalities, Multiple/pathology , Alanine-tRNA Ligase/chemistry , Amino Acid Sequence , Base Sequence , Epilepsy/pathology , Genes, Recessive/genetics , Humans , Infant , Infant, Newborn , Molecular Sequence Data , Mutation/genetics , Peripheral Nervous System Diseases/pathology , Prospective Studies , Sequence Analysis, DNA , Syndrome , United States
15.
Hum Mutat ; 35(11): 1363-71, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25168514

ABSTRACT

Charcot-Marie-Tooth disease type 2D (CMT2D) is an autosomal-dominant axonal peripheral neuropathy characterized by impaired motor and sensory function in the distal extremities. Mutations in the glycyl-tRNA synthetase (GARS) gene cause CMT2D. GARS is a member of the ubiquitously expressed aminoacyl-tRNA synthetase (ARS) family and is responsible for charging tRNA with glycine. To date, 13 GARS mutations have been identified in patients with CMT disease. While functional studies have revealed loss-of-function characteristics, only four GARS mutations have been rigorously studied. Here, we report the functional evaluation of nine CMT-associated GARS mutations in tRNA charging, yeast complementation, and subcellular localization assays. Our results demonstrate that impaired function is a common characteristic of CMT-associated GARS mutations. Additionally, one mutation previously associated with CMT disease (p.Ser581Leu) does not demonstrate impaired function, was identified in the general population, and failed to segregate with disease in two newly identified families with CMT disease. Thus, we propose that this variant is not a disease-causing mutation. Together, our data indicate that impaired function is a key component of GARS-mediated CMT disease and emphasize the need for careful genetic and functional evaluation before implicating a variant in disease onset.


Subject(s)
Genetic Association Studies , Glycine-tRNA Ligase/genetics , Glycine-tRNA Ligase/metabolism , Mutation , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Amino Acid Sequence , Aminoacylation , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Conserved Sequence , DNA Mutational Analysis , Female , Gene Expression , Glycine-tRNA Ligase/chemistry , Humans , Kinetics , Male , Mice , Neurons/metabolism , Pedigree , Protein Transport , Yeasts/genetics , Yeasts/metabolism
16.
Genetics ; 194(2): 447-57, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23608191

ABSTRACT

Mammalian male fertility relies on complex inter- and intracellular signaling during spermatogenesis. Here we describe three alleles of the widely expressed A-kinase anchoring protein 9 (Akap9) gene, all of which cause gametogenic failure and infertility in the absence of marked somatic phenotypes. Akap9 disruption does not affect spindle nucleation or progression of prophase I of meiosis but does inhibit maturation of Sertoli cells, which continue to express the immaturity markers anti-Mullerian hormone and thyroid hormone receptor alpha in adults and fail to express the maturation marker p27(Kip1). Furthermore, gap and tight junctions essential for blood-testis barrier (BTB) organization are disrupted. Connexin43 (Cx43) and zona occludens-1 are improperly localized in Akap9 mutant testes, and Cx43 fails to compartmentalize germ cells near the BTB. These results identify and support a novel reproductive tissue-specific role for Akap9 in the coordinated regulation of Sertoli cells in the testis.


Subject(s)
A Kinase Anchor Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Sertoli Cells/cytology , Spermatogenesis/genetics , A Kinase Anchor Proteins/genetics , Animals , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , Connexin 43/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gap Junctions/ultrastructure , Male , Meiosis/genetics , Mice , Mice, Mutant Strains , Microtubule-Associated Proteins/genetics , Organ Specificity , Protein Transport , Sertoli Cells/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Spindle Apparatus/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein/metabolism
17.
J Natl Cancer Inst ; 103(12): 962-78, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21653923

ABSTRACT

BACKGROUND: Chromosomal translocations generating oncogenic transcription factors are the hallmark of a variety of tumors, including many sarcomas. Ewing sarcoma family of tumors (ESFTs) are characterized by the t(11;22)(q24;q12) translocation that generates the Ewing sarcoma breakpoint region 1 and Friend leukemia virus integration 1 (EWS-FLI1) fusion transcription factor responsible for the highly malignant phenotype of this tumor. Although continued expression of EWS-FLI1 is believed to be critical for ESFT cell survival, a clinically effective small-molecule inhibitor remains elusive likely because EWS-FLI1 is a transcription factor and therefore widely felt to be "undruggable." METHODS: We developed a high-throughput screen to evaluate more than 50 000 compounds for inhibition of EWS-FLI1 activity in TC32 ESFT cells. We used a TC32 cell-based luciferase reporter screen using the EWS-FLI1 downstream target NR0B1 promoter and a gene signature secondary screen to sort and prioritize the compounds. We characterized the lead compound, mithramycin, based on its ability to inhibit EWS-FLI1 activity in vitro using microarray expression profiling, quantitative reverse transcription-polymerase chain reaction, and immunoblot analysis, and in vivo using immunohistochemistry. We studied the impact of this inhibition on cell viability in vitro and on tumor growth in ESFT xenograft models in vivo (n = 15-20 mice per group). All statistical tests were two-sided. RESULTS: Mithramycin inhibited expression of EWS-FLI1 downstream targets at the mRNA and protein levels and decreased the growth of ESFT cells at half maximal inhibitory concentrations between 10 (95% confidence interval [CI] = 8 to 13 nM) and 15 nM (95% CI = 13 to 19 nM). Mithramycin suppressed the growth of two different ESFT xenograft tumors and prolonged the survival of ESFT xenograft-bearing mice by causing a decrease in mean tumor volume. For example, in the TC32 xenograft model, on day 15 of treatment, the mean tumor volume for the mithramycin-treated mice was approximately 3% of the tumor volume observed in the control mice (mithramycin vs control: 69 vs 2388 mm(3), difference = 2319 mm(3), 95% CI = 1766 to 2872 mm(3), P < .001). CONCLUSION: Mithramycin inhibits EWS-FLI1 activity and demonstrates ESFT antitumor activity both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , High-Throughput Screening Assays/methods , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/genetics , Plicamycin/pharmacology , Proto-Oncogene Protein c-fli-1/drug effects , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/drug effects , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , Immunohistochemistry , Mice , Microscopy, Confocal , Protein Array Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/drug therapy , Transcription Factors/genetics , Transcription, Genetic/drug effects , Translocation, Genetic/drug effects , Transplantation, Heterologous
18.
Neoplasia ; 13(2): 145-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21403840

ABSTRACT

ET-743 (trabectedin; Yondelis) is approved in Europe for the treatment of soft tissue sarcomas. Emerging phase 1 and 2 clinical data have shown high response rates in myxoid liposarcoma in part owing to the inhibition of the FUS-CHOP transcription factor. In this report, we show that modulation of specific oncogenic transcription factors by ET-743 may extend to other tumor types. We demonstrate that, among a panel of pediatric sarcomas, Ewing sarcoma family of tumors (ESFTs) cell lines bearing the EWS-FLI1 transcription factor are the most sensitive to treatment with ET-743 compared with osteosarcoma, rhabdomyosarcoma, and synovial sarcoma. We show that ET-743 reverses a gene signature of induced downstream targets of EWS-FLI1 in two different ESFT cell lines (P = .001). In addition, ET-743 directly suppresses the promoter activity of a known EWS-FLI1 downstream target NR0B1 luciferase reporter construct without changing the activity of a constitutively active control in ESFT cells. Furthermore, the effect is specific to EWS-FLI1, as forced expression of EWS-FLI1 in a cell type that normally lacks this fusion protein, HT1080 cells, induces the same NR0B1 promoter, but this activation is completely blocked by ET-743 treatment. Finally, we used gene set enrichment analysis to confirm that other mechanisms of ET-743 are active in ESFT cells. These results suggest a particular role for ET-743 in the treatment of translocation-positive tumors. In addition, the modulation of EWS-FLI1 makes it a novel targeting agent for ESFT and suggests that further development of this compound for the treatment of ESFT is warranted.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Bone Neoplasms/drug therapy , Dioxoles/therapeutic use , Oncogene Proteins, Fusion/antagonists & inhibitors , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , RNA-Binding Protein EWS/antagonists & inhibitors , Sarcoma, Ewing/drug therapy , Tetrahydroisoquinolines/therapeutic use , Cell Line, Tumor , Child , DAX-1 Orphan Nuclear Receptor/drug effects , DAX-1 Orphan Nuclear Receptor/genetics , Europe , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic/drug effects , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , RNA-Binding Protein FUS/drug effects , Sarcoma/drug therapy , Trabectedin , Transcription Factor CHOP/drug effects
19.
PLoS Genet ; 3(8): e139, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17784788

ABSTRACT

Crossing over during meiotic prophase I is required for sexual reproduction in mice and contributes to genome-wide genetic diversity. Here we report on the characterization of an N-ethyl-N-nitrosourea-induced, recessive allele called mei4, which causes sterility in both sexes owing to meiotic defects. In mutant spermatocytes, chromosomes fail to congress properly at the metaphase plate, leading to arrest and apoptosis before the first meiotic division. Mutant oocytes have a similar chromosomal phenotype but in vitro can undergo meiotic divisions and fertilization before arresting. During late meiotic prophase in mei4 mutant males, absence of cyclin dependent kinase 2 and mismatch repair protein association from chromosome cores is correlated with the premature separation of bivalents at diplonema owing to lack of chiasmata. We have identified the causative mutation, a transversion in the 5' splice donor site of exon 1 in the mouse ortholog of Human Enhancer of Invasion 10 (Hei10; also known as Gm288 in mouse and CCNB1IP1 in human), a putative B-type cyclin E3 ubiquitin ligase. Importantly, orthologs of Hei10 are found exclusively in deuterostomes and not in more ancestral protostomes such as yeast, worms, or flies. The cloning and characterization of the mei4 allele of Hei10 demonstrates a novel link between cell cycle regulation and mismatch repair during prophase I.


Subject(s)
Cell Cycle Proteins/genetics , Crossing Over, Genetic/genetics , Meiotic Prophase I/genetics , Mutation , Ubiquitin-Protein Ligases/genetics , Adaptor Proteins, Signal Transducing , Alleles , Animals , Base Pair Mismatch/genetics , Cattle , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase 2/deficiency , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Recombination, Genetic , Ubiquitin-Protein Ligases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...