Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
J Vet Cardiol ; 40: 119-125, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34144877

ABSTRACT

INTRODUCTION: The study objectives were to determine the prevalence and geographic distribution of a dilated cardiomyopathy (DCM)-associated RNA-binding motif protein 20 (RBM20) variant in canine DNA samples submitted for testing and to evaluate the influence of the genotype on cardiac phenotype and lifespan. ANIMALS: Samples from 2136 dogs including 1834 Standard Schnauzers (SSNZ), 266 Giant Schnauzers (GSNZ), and 36 dogs of other breeds. METHODS: The University of Missouri Canine Genetics Laboratory's sample-accession spreadsheet and Orthopedic Foundation for Animals' database were retrospectively reviewed for samples submitted for RBM20 genotyping from November, 2013, through May, 2018. Data analyzed included breed, date of birth, RBM20 genotype (homozygous wild-type, heterozygous variant [HET], or homozygous variant [HOM]), geographic origin of submission, pedigree, cardiac phenotype, and date of death or current age if alive. RESULTS AND DISCUSSION: The RBM20 variant was only detected in SSNZ and GSNZ. A total of 389 SSNZ were variant-positive (prevalence = 21.2%), with 361 HET (19.7%) and 28 HOM (1.5%). Of the HOM SSNZ, DCM was confirmed in 26 of 28 (92.9%), with the remainder lost to follow-up. The median lifespan of HOM SSNZ (3.06 years) was significantly shorter than that for HET (15.11 years) and wild-type (15.18 years) SSNZ. Twenty-six GSNZ were variant-positive (prevalence = 9.8%), with 23 HET (8.6%) and three HOM (1.1%). Nine GSNZ belonged to one family, including the three HOM GSNZ that all had DCM. CONCLUSIONS: The HOM genotype is associated with DCM and premature death in SSNZ and GSNZ.


Subject(s)
Cardiomyopathy, Dilated , Dog Diseases , Animals , Cardiomyopathy, Dilated/epidemiology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/veterinary , Dog Diseases/epidemiology , Dog Diseases/genetics , Dogs , Genotype , Longevity , Prevalence , RNA-Binding Motifs , Retrospective Studies
2.
Sci Rep ; 10(1): 21399, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293645

ABSTRACT

Genotype-phenotype correlations of humans and dogs with hereditary methemoglobinemia are not yet well characterized. We determined total hemoglobin and methemoglobin (MetHb) concentrations, cytochrome b5 reductase (CYB5R) enzyme activities, genotypes, and clinical signs in 30 dogs with persistent cyanosis without cardiopulmonary disease. Erythrocytic CYB5R enzyme activities were low in all dogs assayed. Owner-reported quality of life ranged from subclinical to occasional exertional syncope. Two previously reported and two novel CYB5R3 missense variants were identified among the methemoglobinemic cohort and were predicted to impair enzyme function. Two variants were recurrent: a homozygous Ile194Leu substitution was found in Pomeranians and other small dogs, and a homozygous Arg219Pro change occurred predominately in pit bull terriers. The other two variants were Thr202Ala and Gly76Ser substitutions in single dogs. Of the two common CYB5R3 genotypes, Arg219Pro was associated with a more severe metabolic phenotype. We conclude that CYB5R3 deficiency is the predominate cause of canine hereditary methemoglobinemia. Although this finding is unlikely to alter the clinical approach to hereditary methemoglobinemia in dogs, it demonstrates the possibility of how genotype-phenotype cohort analysis might facilitate precision medicine in the future in veterinary medicine.


Subject(s)
Cytochrome-B(5) Reductase/genetics , Methemoglobinemia/congenital , Mutation, Missense , Amino Acid Substitution , Animals , Cytochrome-B(5) Reductase/deficiency , Dogs , Female , Genetic Predisposition to Disease , Hemoglobins/metabolism , Male , Methemoglobin/metabolism , Methemoglobinemia/genetics , Methemoglobinemia/metabolism , Prospective Studies
3.
Anim Genet ; 50(5): 543-545, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31294848

ABSTRACT

An eight-week old Doberman Pinscher was diagnosed with Ehlers Danlos syndrome based on the dog's hyper-mobile carpal, tarsal and stifle joints and abnormal skin. The skin was loose and hyper-elastic with several wounds and large atrophic scars. The dog was euthanized after a severe degloving injury from minimal trauma. A whole-genome sequence, generated with DNA from the dog's blood, contained a rare, homozygous C-to-T transition at position 2408978 on chromosome 11. This transition is predicted to alter the ADAMTS2 transcript (ADAMTS2:c.769C>T) and encode a nonsense mutation (p.Arg257Ter). Biallelic ADAMTS2 mutations have caused a type of Ehlers Danlos syndrome known as dermatosparaxis in other species.


Subject(s)
ADAMTS Proteins/genetics , Dog Diseases/genetics , Ehlers-Danlos Syndrome/veterinary , Skin Diseases/veterinary , Animals , Dogs , Ehlers-Danlos Syndrome/genetics , Skin Diseases/genetics
4.
J Vet Intern Med ; 31(6): 1860-1865, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28963729

ABSTRACT

A juvenile male mixed breed dog was presented for lethargy, exercise intolerance, and aggression when touched on the head. Cyanosis, tachycardia, and tachypnea were observed and persisted during oxygen supplementation. Arterial blood gas analysis by co-oximetry identified an increased methemoglobin concentration (27%; normal, <2%) with normal arterial oxygen tension. The methemoglobinemia and associated clinical signs resolved after administration of methylene blue (1 mg/kg) IV, and the dog was discharged. The affected dog's whole-genome sequence contained 2 potentially causal heterozygous CYB5R3 missense mutations suggesting that cytochrome b5 reductase deficiency was responsible for the methemoglobinemia. This hypothesis was confirmed by enzyme analysis that identified cytochrome b5 reductase activity in the affected dog's erythrocytes to only approximately 6% of that in a control sample. Clinical signs recurred 11 days after discharge but normalized and the methemoglobin concentration decreased with methylene blue administration PO (1.5 mg/kg, initially daily and then every other day).


Subject(s)
Cytochrome-B(5) Reductase/deficiency , Dog Diseases/genetics , Methemoglobinemia/veterinary , Methylene Blue/therapeutic use , Animals , Blood Gas Analysis/veterinary , Cytochrome-B(5) Reductase/genetics , Dog Diseases/drug therapy , Dogs , Erythrocytes/enzymology , Male , Methemoglobinemia/drug therapy , Methemoglobinemia/genetics , Methylene Blue/administration & dosage , Mutation, Missense , Whole Genome Sequencing/veterinary
5.
J Vet Intern Med ; 31(5): 1520-1526, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28833537

ABSTRACT

Consistent with a tentative diagnosis of neuronal ceroid lipofuscinosis (NCL), autofluorescent cytoplasmic storage bodies were found in neurons from the brains of 2 related Shiba Inu dogs with a young-adult onset, progressive neurodegenerative disease. Unexpectedly, no potentially causal NCL-related variants were identified in a whole-genome sequence generated with DNA from 1 of the affected dogs. Instead, the whole-genome sequence contained a homozygous 3 base pair (bp) deletion in a coding region of HEXB. The other affected dog also was homozygous for this 3-bp deletion. Mutations in the human HEXB ortholog cause Sandhoff disease, a type of GM2 gangliosidosis. Thin-layer chromatography confirmed that GM2 ganglioside had accumulated in an affected Shiba Inu brain. Enzymatic analysis confirmed that the GM2 gangliosidosis resulted from a deficiency in the HEXB encoded protein and not from a deficiency in products from HEXA or GM2A, which are known alternative causes of GM2 gangliosidosis. We conclude that the homozygous 3-bp deletion in HEXB is the likely cause of the Shiba Inu neurodegenerative disease and that whole-genome sequencing can lead to the early identification of potentially disease-causing DNA variants thereby refocusing subsequent diagnostic analyses toward confirming or refuting candidate variant causality.


Subject(s)
Dog Diseases/genetics , Gangliosidoses, GM2/veterinary , Gene Deletion , beta-Hexosaminidase beta Chain/genetics , Animals , Dog Diseases/pathology , Dogs , Female , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/pathology , Homozygote , Microscopy, Electron/veterinary
6.
J Vet Intern Med ; 31(1): 149-157, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28008682

ABSTRACT

A 10-month-old spayed female Cane Corso dog was evaluated after a 2-month history of progressive blindness, ataxia, and lethargy. Neurologic examination abnormalities indicated a multifocal lesion with primarily cerebral and cerebellar signs. Clinical worsening resulted in humane euthanasia. On necropsy, there was marked astrogliosis throughout white matter tracts of the cerebrum, most prominently in the corpus callosum. In the cerebral cortex and midbrain, most neurons contained large amounts of autofluorescent storage material in the perinuclear area of the cells. Cerebellar storage material was present in the Purkinje cells, granular cell layer, and perinuclear regions of neurons in the deep nuclei. Neuronal ceroid lipofuscinosis (NCL) was diagnosed. Whole genome sequencing identified a PPT1c.124 + 1G>A splice donor mutation. This nonreference assembly allele was homozygous in the affected dog, has not previously been reported in dbSNP, and was absent from the whole genome sequences of 45 control dogs and 31 unaffected Cane Corsos. Our findings indicate a novel mutation causing the CLN1 form of NCL in a previously unreported dog breed. A canine model for CLN1 disease could provide an opportunity for therapeutic advancement, benefiting both humans and dogs with this disorder.


Subject(s)
Dog Diseases/diagnosis , Neuronal Ceroid-Lipofuscinoses/veterinary , Animals , Dog Diseases/genetics , Dogs , Female , Frameshift Mutation/genetics , Magnetic Resonance Imaging/veterinary , Neuronal Ceroid-Lipofuscinoses/diagnosis , Neuronal Ceroid-Lipofuscinoses/diagnostic imaging , Neuronal Ceroid-Lipofuscinoses/genetics
7.
J Vet Intern Med ; 30(4): 1149-58, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27203721

ABSTRACT

BACKGROUND: Neuronal ceroid lipofuscinosis (NCL), a fatal neurodegenerative disease, has been diagnosed in young adult Australian Cattle Dogs. OBJECTIVE: Characterize the Australian Cattle Dog form of NCL and determine its molecular genetic cause. ANIMALS: Tissues from 4 Australian Cattle Dogs with NCL-like signs and buccal swabs from both parents of a fifth affected breed member. Archived DNA samples from 712 individual dogs were genotyped. METHODS: Tissues were examined by fluorescence, electron, and immunohistochemical microscopy. A whole-genome sequence was generated for 1 affected dog. A TaqMan allelic discrimination assay was used for genotyping. RESULTS: The accumulation of autofluorescent cytoplasmic storage material with characteristic ultrastructure in tissues from the 4 affected dogs supported a diagnosis of NCL. The whole-genome sequence contained a homozygous nonsense mutation: CLN5:c.619C>T. All 4 DNA samples from clinically affected dogs tested homozygous for the variant allele. Both parents of the fifth affected dog were heterozygotes. Archived DNA samples from 346 Australian Cattle Dogs, 188 Border Collies, and 177 dogs of other breeds were homozygous for the reference allele. One archived Australian Cattle Dog sample was from a heterozygote. CONCLUSIONS AND CLINICAL IMPORTANCE: The homozygous CLN5 nonsense is almost certainly causal because the same mutation previously had been reported to cause a similar form of NCL in Border Collies. Identification of the molecular genetic cause of Australian Cattle Dog NCL will allow the use of DNA tests to confirm the diagnosis of NCL in this breed.


Subject(s)
Dog Diseases/genetics , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/veterinary , Animals , Codon, Nonsense , Dogs , Female , Genetic Predisposition to Disease , Male , Neuronal Ceroid-Lipofuscinoses/genetics , Pedigree
8.
J Vet Intern Med ; 30(3): 813-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26968732

ABSTRACT

BACKGROUND: A variety of presumed hereditary, neurologic diseases have been reported in young Rottweilers. Overlapping ages of onset and clinical signs have made antemortem diagnosis difficult. One of these diseases, neuronal vacuolation and spinocerebellar degeneration (NVSD) shares clinical and histological features with polyneuropathy with ocular abnormalities and neuronal vacuolation (POANV), a recently described hereditary disease in Black Russian Terriers (BRTs). Dogs with POANV harbor mutations in RAB3GAP1 which codes for a protein involved in membrane trafficking. HYPOTHESIS: Rottweilers with NVSD will be homozygous for the RAB3GAP1:c.743delC allele associated with POANV in BRTs. ANIMALS: Eight Rottweilers with NVSD confirmed at necropsy, 128 Rottweilers without early onset neurologic signs, and 468 randomly selected dogs from 169 other breeds. METHODS: Retrospective case-control study. Dogs were genotyped for the RAB3GAP1:c.743delC allele with an allelic discrimination assay. RESULTS: All 8 NVSD-affected dogs were homozygous for the RAB3GAP1:c.743delC allele while the 128 NVSD-free Rottweilers were either homozygous for the reference allele (n = 105) or heterozygous (n = 23) and the 468 genotyped dogs from other breeds were all homozygous for the reference allele. CONCLUSIONS AND CLINICAL IMPORTANCE: The RAB3GAP1:c.743delC mutation is associated with a similar phenotype in Rottweilers and BRTs. Identification of the mutation permits a DNA test that can aid in the diagnosis of NVSD and identify carriers of the trait so that breeders can avoid producing affected dogs. Disruption of membrane trafficking could explain the neuronal vacuolation seen in NVSD and other spongiform encephalopathies.


Subject(s)
Dog Diseases/genetics , Spinocerebellar Degenerations/veterinary , rab3 GTP-Binding Proteins/genetics , Animals , Dog Diseases/pathology , Dogs , Genotype , Mutation , Neurons/pathology , Polyneuropathies/genetics , Polyneuropathies/pathology , Polyneuropathies/veterinary , Retrospective Studies , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/pathology
9.
Mol Genet Metab ; 115(2-3): 101-9, 2015.
Article in English | MEDLINE | ID: mdl-25934231

ABSTRACT

We studied a recessive, progressive neurodegenerative disease occurring in Golden Retriever siblings with an onset of signs at 15 months of age. As the disease progressed these signs included ataxia, anxiety, pacing and circling, tremors, aggression, visual impairment and localized and generalized seizures. A whole genome sequence, generated with DNA from one affected dog, contained a plausibly causal homozygous mutation: CLN5:c.934_935delAG. This mutation was predicted to produce a frameshift and premature termination codon and encode a protein variant, CLN5:p.E312Vfs*6, which would lack 39 C-terminal amino acids. Eighteen DNA samples from the Golden Retriever family members were genotyped at CLN5:c.934_935delAG. Three clinically affected dogs were homozygous for the deletion allele; whereas, the clinically normal family members were either heterozygotes (n = 11) or homozygous for the reference allele (n = 4). Among archived Golden Retrievers DNA samples with incomplete clinical records that were also genotyped at the CLN5:c.934_935delAG variant, 1053 of 1062 were homozygous for the reference allele, 8 were heterozygotes and one was a deletion-allele homozygote. When contacted, the owner of this homozygote indicated that their dog had been euthanized because of a neurologic disease that progressed similarly to that of the affected Golden Retriever siblings. We have collected and stored semen from a heterozygous Golden Retriever, thereby preserving an opportunity for us or others to establish a colony of CLN5-deficient dogs.


Subject(s)
Dog Diseases/genetics , Frameshift Mutation , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/veterinary , Sequence Deletion , Animals , Base Sequence , Dogs , Homozygote , Neuronal Ceroid-Lipofuscinoses/genetics , Sequence Analysis, DNA
10.
Curr Alzheimer Res ; 12(1): 22-31, 2015.
Article in English | MEDLINE | ID: mdl-25523423

ABSTRACT

BACKGROUND: Previous studies showed that Bryostatin-1, a potent PKC modulator and alphasecretase activator, can improve cognition in models of Alzheimer's disease (AD) with chronic (>10 weeks), intraperitoneal (i.p.) administration of the drug. We compared learning and spatial memory in the APPswe, PSEN1dE985Dbo (APP/PS1) mouse model of AD and studied the ability of acute intraperitoneal and oral Bryostatin-1 to reverse cognitive deficits in this model. Compared to wild-type (WT) mice, APP/PS1 mice showed significant delays in learning the location of a submerged platform in the Morris water maze. Bryostatin-1 was administered over a 2-week course prior to and during water maze testing. RESULTS: Acute i.p. Bryostatin-1 administration did not improve latency to escape but oral Bryostatin-1 significantly improved memory (measured by a reduction in latency to escape). This benefit of oral Bryostatin-1 administration was most apparent during the first 3 days of testing. These findings show that: 1) Bryostatin-1 is orally active in models of learning and memory, 2) this effect can be produced in less than 2 weeks and 3) this effect is not seen with i.p. administration. We conclude that oral Bryostatin-1 represents a novel, potent and long-acting memory enhancer with future clinical applications in the treatment of human AD.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alzheimer Disease/complications , Bryostatins/therapeutic use , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Administration, Oral , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Escape Reaction/drug effects , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Reaction Time/drug effects
11.
Opt Express ; 22(11): 13600-15, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24921554

ABSTRACT

We demonstrate logic functionalities in a high-speed all-optical logic circuit based on differential Mach-Zehnder interferometers with semiconductor optical amplifiers as the nonlinear optical elements. The circuit, implemented by hybrid integration of the semiconductor optical amplifiers on a planar lightwave circuit platform fabricated in silica glass, can be flexibly configured to realize a variety of Boolean logic gates. We present both simulations and experimental demonstrations of cascaded all-optical operations for 80-Gb/s on-off keyed data.

12.
J Vet Intern Med ; 28(3): 871-7, 2014.
Article in English | MEDLINE | ID: mdl-24708069

ABSTRACT

BACKGROUND: Juvenile-onset spinocerebellar ataxia has been recognized in Jack Russell Terriers and related Russell group terriers (RGTs) for over 40 years. Ataxia occurs with varying combinations of myokymia, seizures, and other signs of neurologic disease. More than 1 form of the disease has been suspected. HYPOTHESIS/OBJECTIVES: The objective was to identify the mutation causing the spinocerebellar ataxia associated with myokymia, seizures, or both and distinguish the phenotype from other ataxias in the RGTs. ANIMALS: DNA samples from 16 RGTs with spinocerebellar ataxia beginning from 2 to 12 months of age, 640 control RGTs, and 383 dogs from 144 other breeds along with the medical records of affected dogs were studied. METHODS: This case-control study compared the frequencies of a KCNJ10 allele in RGTs with spinocerebellar ataxia versus control RGTs. This allele was identified in a whole-genome sequence of a single RGT with spinocerebellar ataxia and myokymia by comparison to whole-genome sequences from 81 other canids that were normal or had other diseases. RESULTS: A missense mutation in the gene coding for the inwardly rectifying potassium channel Kir4.1 (KCNJ10:c.627C>G) was significantly (P < .001) associated with the disease. Dogs homozygous for the mutant allele all had spinocerebellar ataxia with varying combinations of myokymia and seizures. CONCLUSIONS AND CLINICAL IMPORTANCE: Identification of the KCNJ10 mutation in dogs with spinocerebellar ataxia with myokymia, seizures, or both clarifies the multiple forms of ataxia seen in these breeds and provides a DNA test to identify carriers.


Subject(s)
Dog Diseases/genetics , Mutation, Missense/genetics , Myokymia/veterinary , Potassium Channels, Inwardly Rectifying/genetics , Seizures/veterinary , Spinocerebellar Ataxias/veterinary , Alleles , Animals , Case-Control Studies , Dogs , Female , Genetic Predisposition to Disease/genetics , Homozygote , Male , Myokymia/genetics , Seizures/genetics , Spinocerebellar Ataxias/genetics
13.
J Vet Intern Med ; 28(2): 515-21, 2014.
Article in English | MEDLINE | ID: mdl-24524809

ABSTRACT

BACKGROUND: Previous reports associated 2 mutant SOD1 alleles (SOD1:c.118A and SOD1:c.52T) with degenerative myelopathy in 6 canine breeds. The distribution of these alleles in other breeds has not been reported. OBJECTIVE: To describe the distribution of SOD1:c.118A and SOD1:c.52T in 222 breeds. ANIMALS: DNA from 33,747 dogs was genotyped at SOD1:c.118, SOD1:c.52, or both. Spinal cord sections from 249 of these dogs were examined. METHODS: Retrospective analysis of 35,359 previously determined genotypes at SOD1:c.118G>A or SOD1:c.52A>T and prospective survey to update the clinical status of a subset of dogs from which samples were obtained with a relatively low ascertainment bias. RESULTS: The SOD1:c.118A allele was found in cross-bred dogs and in 124 different canine breeds whereas the SOD1:c.52T allele was only found in Bernese Mountain Dogs. Most of the dogs with histopathologically confirmed degenerative myelopathy were SOD1:c.118A homozygotes, but 8 dogs with histopathologically confirmed degenerative myelopathy were SOD1:c.118A/G heterozygotes and had no other sequence variants in their SOD1 amino acid coding regions. The updated clinical conditions of dogs from which samples were obtained with a relatively low ascertainment bias suggest that SOD1:c.118A homozygotes are at a much higher risk of developing degenerative myelopathy than are SOD1:c.118A/G heterozygotes. CONCLUSIONS AND CLINICAL IMPORTANCE: We conclude that the SOD1:c.118A allele is widespread and common among privately owned dogs whereas the SOD1:c.52T allele is rare and appears to be limited to Bernese Mountain Dogs. We also conclude that breeding to avoid the production of SOD1:c.118A homozygotes is a rational strategy.


Subject(s)
Dog Diseases/genetics , Muscular Atrophy, Spinal/veterinary , Superoxide Dismutase/genetics , Alleles , Animals , Dogs/genetics , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Homozygote , Muscular Atrophy, Spinal/genetics , Mutation, Missense , Species Specificity
14.
Vet Pathol ; 51(4): 796-804, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24026940

ABSTRACT

Three young domestic shorthair cats were presented for necropsy with similar histories of slowly progressive visual dysfunction and neurologic deficits. Macroscopic examination of each cat revealed cerebral and cerebellar atrophy, dilated lateral ventricles, and slight brown discoloration of the gray matter. Histologically, there was bilateral loss of neurons within the limbic, motor, somatosensory, visual, and, to a lesser extent, vestibular systems with extensive astrogliosis in the affected regions of all 3 cases. Many remaining neurons and glial cells throughout the entire central nervous system were distended by pale yellow to eosinophilic, autofluorescent cytoplasmic inclusions with ultrastructural appearances typical of neuronal ceroid-lipofuscinoses (NCLs). Differences in clinical presentation and neurological lesions suggest that the 3 cats may have had different variants of NCL. Molecular genetic characterization in the 1 cat from which DNA was available did not reveal any plausible disease-causing mutations of the CLN1 (PPT1), CLN3, CLN5, CLN8, and CLN10 (CTSD) genes. Further investigations will be required to identify the mutations responsible for NCLs in cats.


Subject(s)
Cat Diseases/pathology , Nervous System/pathology , Neuronal Ceroid-Lipofuscinoses/veterinary , Animals , Atrophy/pathology , Atrophy/veterinary , Cats , DNA Mutational Analysis/veterinary , Fatal Outcome , Histological Techniques/veterinary , Immunohistochemistry/veterinary , Minnesota , Neuronal Ceroid-Lipofuscinoses/pathology
15.
Epidemiol Infect ; 140(2): 283-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21554779

ABSTRACT

Routine laboratory testing may not detect non-O157 Shiga toxin-producing Escherichia coli (STEC) reliably. Active clinical, epidemiological, environmental health, and laboratory collaboration probably influence successful detection and study of non-O157 STEC infection. We summarized two outbreak investigations in which such coordinated efforts identified non-O157 STEC disease and led to effective control measures. Outbreak 1 involved illness associated with consuming unpasteurized apple cider from a local orchard. Public health personnel were notified by a local hospital; stool specimens from ill persons contained O111 STEC. Outbreak 2 involved bloody diarrhoea at a correctional facility. Public health personnel were notified by the facility infection control officer; O45 STEC was the implicated agent. These reports highlight the ability of non-O157 STEC to cause outbreaks and demonstrate that a coordinated effort by clinicians, infection-control practitioners, clinical diagnostic laboratorians, and public health personnel can lead to effective identification, investigation, and prevention of non-O157 STEC disease.


Subject(s)
Diarrhea/epidemiology , Diarrhea/microbiology , Disease Outbreaks , Epidemiologic Methods , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Adult , Animals , Cattle , Diarrhea/diagnosis , Escherichia coli Infections/diagnosis , Escherichia coli Infections/transmission , Feces/microbiology , Food Microbiology , Humans , Immunoenzyme Techniques , Incidence , New York , Real-Time Polymerase Chain Reaction , Shiga Toxin 1/analysis , Shiga Toxin 1/genetics , Shiga Toxin 2/analysis , Shiga Toxin 2/genetics , Shiga-Toxigenic Escherichia coli/genetics
17.
J Vet Intern Med ; 25(2): 267-72, 2011.
Article in English | MEDLINE | ID: mdl-21281350

ABSTRACT

BACKGROUND: Bandera's neonatal ataxia (BNAt) is an autosomal recessive cerebellar ataxia that affects members of the Coton de Tulear dog breed. OBJECTIVE: To identify the mutation that causes BNAt. ANIMALS: The study involved DNA from 112 Cotons de Tulear (including 15 puppies with signs of BNAt) and 87 DNA samples from dogs of 12 other breeds. METHODS: The BNAt locus was mapped with a genome-wide association study (GWAS). The coding exons of positional candidate gene GRM1, which encodes metabotropic glutamate receptor 1, were polymerase chain reaction (PCR)-amplified and resequenced. A 3-primer PCR assay was used to genotype individual dogs for a truncated retrotransposon inserted into exon 8 of GRM1. RESULTS: The GWAS indicated that the BNAt locus was in a canine chromosome 1 region that contained candidate gene GRM1. Resequencing this gene from BNAt-affected puppies indicated that exon 8 was interrupted by the insertion of a 5'-truncated retrotransposon. All 15 BNAt-affected puppies were homozygous for the insert, whereas all other Cotons de Tulear were heterozygotes (n = 43) or homozygous (n = 54) for the ancestral allele. None of the 87 dogs from 12 other breeds had the insertion allele. CONCLUSIONS AND CLINICAL IMPORTANCE: BNAt is caused by a retrotransposon inserted into exon 8 of GRM1. A DNA test for the GRM1 retrotransposon insert can be used for genetic counseling and to confirm the diagnosis of BNAt.


Subject(s)
Cerebellar Ataxia/veterinary , Dog Diseases/genetics , Mutation , Receptors, Metabotropic Glutamate/genetics , Age of Onset , Animals , Cerebellar Ataxia/genetics , DNA Mutational Analysis/veterinary , DNA Primers/genetics , Dogs , Exons , Female , Genome-Wide Association Study , Genotype , Heterozygote , Homozygote , Male , Mutagenesis, Insertional , Open Reading Frames , Pedigree , Retroelements
19.
Article in English | MEDLINE | ID: mdl-19717540

ABSTRACT

The domestic dog offers a remarkable opportunity to disentangle the genetics of complex phenotypes. Here, we explore a locus, previously identified in the Portuguese water dog (PWD), associated with PC2, a morphological principal component characterized as leg width versus leg length. The locus was initially mapped to a region of 26 Mb on canine chromosome 12 (CFA12) following a genome-wide scan. Subsequent and extensive genotyping of single-nucleotide polymorphisms (SNPs) and haplotype analysis in both the PWD and selected breeds representing phenotypic extremes of PC2 reduced the region from 26 Mb to 500 kb. The proximity of the critical interval to two collagen genes suggests that the phenotype may be controlled by cis-acting mechanisms.


Subject(s)
Dogs/anatomy & histology , Dogs/genetics , Extremities/anatomy & histology , Animals , Chromosome Mapping , Genetic Association Studies , Genome-Wide Association Study , Haplotypes , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci
SELECTION OF CITATIONS
SEARCH DETAIL
...