Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 11(5)2020 04 28.
Article in English | MEDLINE | ID: mdl-32354065

ABSTRACT

A 4-month-old female Irish Terrier presented with a well demarcated ulcerative and crusting lesion in the right ear canal. Histological analysis revealed epidermal hyperplasia with severe acantholysis affecting all suprabasal layers of the epidermis, which prompted a presumptive diagnosis of canine Darier disease. The lesion was successfully treated by repeated laser ablation of the affected epidermis. Over the course of three years, the dog additionally developed three dermal nodules of up to 4 cm in diameter that were excised and healed without complications. Histology of the excised tissue revealed multiple infundibular cysts extending from the upper dermis to the subcutis. The cysts were lined by squamous epithelium, which presented with abundant acantholysis of suprabasal keratinocytes. Infundibular cysts represent a novel finding not previously reported in Darier patients. Whole genome sequencing of the affected dog was performed, and the functional candidate genes for Darier disease (ATP2A2) and Hailey-Hailey disease (ATP2C1) were investigated. The analysis revealed a heterozygous SINE insertion into the ATP2A2 gene, at the end of intron 14, close to the boundary of exon 15. Analysis of the ATP2A2 mRNA from skin of the affected dog demonstrated a splicing defect and marked allelic imbalance, suggesting nonsense-mediated decay of the resulting aberrant transcripts. As Darier disease in humans is caused by haploinsufficiency of ATP2A2, our genetic findings are in agreement with the clinical and histopathological data and support the diagnosis of canine Darier disease.


Subject(s)
Calcium-Transporting ATPases/genetics , Darier Disease/genetics , Pemphigus, Benign Familial/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Acantholysis/genetics , Acantholysis/pathology , Animals , Darier Disease/pathology , Darier Disease/veterinary , Dog Diseases/genetics , Dog Diseases/pathology , Dogs , Ear Canal/metabolism , Ear Canal/pathology , Epidermis/metabolism , Epidermis/pathology , Female , Haploinsufficiency/genetics , Heterozygote , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Pemphigus, Benign Familial/pathology , Pemphigus, Benign Familial/veterinary , Skin/metabolism , Skin/pathology
2.
Genes (Basel) ; 10(10)2019 09 21.
Article in English | MEDLINE | ID: mdl-31546637

ABSTRACT

The Ehlers-Danlos syndromes (EDS) are a heterogeneous group of heritable disorders affecting connective tissues. The mutations causing the various forms of EDS in humans are well characterized, but the genetic mutations causing EDS-like clinical pathology in dogs are not known, thus hampering accurate clinical diagnosis. Clinical analysis of two independent cases of skin hyperextensibility and fragility, one with pronounced joint hypermobility was suggestive of EDS. Whole-genome sequencing revealed de novo mutations of COL5A1 in both cases, confirming the diagnosis of the classical form of EDS. The heterozygous COL5A1 p.Gly1013ValfsTer260 mutation characterized in case 1 introduced a premature termination codon and would be expected to result in α1(V) mRNA nonsense-mediated mRNA decay and collagen V haploinsufficiency. While mRNA was not available from this dog, ultrastructural analysis of the dermis demonstrated variability in collagen fibril diameter and the presence of collagen aggregates, termed 'collagen cauliflowers', consistent with COL5A1 mutations underlying classical EDS. In the second case, DNA sequencing demonstrated a p.Gly1571Arg missense variant in the COL5A1 gene. While samples were not available for further analysis, such a glycine substitution would be expected to destabilize the strict molecular structure of the collagen V triple helix and thus affect protein stability and/or integration of the mutant collagen into the collagen V/collagen I heterotypic dermal fibrils. This is the first report of genetic variants in the COL5A1 gene causing the clinical presentation of EDS in dogs. These data provided further evidence of the important role of collagen V in dermal collagen fibrillogenesis. Importantly, from the clinical perspective, we showed the utility of DNA sequencing, combined with the established clinical criteria, in the accurate diagnosis of EDS in dogs.


Subject(s)
Collagen Type V/genetics , Dog Diseases/genetics , Dogs/genetics , Ehlers-Danlos Syndrome/genetics , Animals , Ehlers-Danlos Syndrome/veterinary , Genetic Variation , Male , Microscopy, Electron, Transmission , Mutation , Skin/metabolism , Skin/pathology , Skin/ultrastructure
3.
Genes (Basel) ; 10(5)2019 05 21.
Article in English | MEDLINE | ID: mdl-31117272

ABSTRACT

Canine progressive retinal atrophies (PRA) are genetically heterogeneous diseases characterized by retinal degeneration and subsequent blindness. PRAs are untreatable and affect multiple dog breeds, significantly impacting welfare. Three out of seven Giant Schnauzer (GS) littermates presented with PRA around four years of age. We sought to identify the causal variant to improve our understanding of the aetiology of this form of PRA and to enable development of a DNA test. Whole genome sequencing of two PRA-affected full-siblings and both unaffected parents was performed. Variants were filtered based on those segregating appropriately for an autosomal recessive disorder and predicted to be deleterious. Successive filtering against 568 canine genomes identified a single nucleotide variant in the gene encoding NECAP endocytosis associated 1 (NECAP1): c.544G>A (p.Gly182Arg). Five thousand one hundred and thirty canids of 175 breeds, 10 cross-breeds and 3 wolves were genotyped for c.544G>A. Only the three PRA-affected GS were homozygous (allele frequency in GS, excluding proband family = 0.015). In addition, we identified heterozygotes belonging to Spitz and Dachshund varieties, demonstrating c.544G>A segregates in other breeds of German origin. This study, in parallel with the known retinal expression and role of NECAP1 in clathrin mediated endocytosis (CME) in synapses, presents NECAP1 as a novel candidate gene for retinal degeneration in dogs and other species.


Subject(s)
Adaptor Protein Complex alpha Subunits/genetics , Dog Diseases/genetics , Retina/pathology , Retinal Degeneration/genetics , Animals , Atrophy/genetics , Atrophy/pathology , Breeding , Dog Diseases/pathology , Dogs , Endocytosis/genetics , Frameshift Mutation , Genome-Wide Association Study , Genotype , Humans , Retinal Degeneration/pathology , Synapses/genetics , Synapses/pathology , Whole Genome Sequencing
5.
PLoS One ; 14(2): e0212645, 2019.
Article in English | MEDLINE | ID: mdl-30794648

ABSTRACT

Bald thigh syndrome is a common hair loss disorder in sighthounds. Numerous possible causes, including environmental conditions, trauma, stress, endocrinopathies and genetic components have been proposed, but only endocrinopathies have been ruled out scientifically. The overall goal of our study was to identify the cause of bald thigh syndrome and the pathological changes associated with it. We approached this aim by comparing skin biopsies and hair shafts of affected and control dogs microscopically as well as by applying high-throughput technologies such as genomics, transcriptomics and proteomics. While the histology is rather unspecific in most cases, trichogram analysis and scanning electron microscopy revealed severe structural abnormalities in hair shafts of affected dogs. This finding is supported by the results of the transcriptomic and proteomic profiling where genes and proteins important for differentiation of the inner root sheath and the assembly of a proper hair shaft were downregulated. Transcriptome profiling revealed a downregulation of genes encoding 23 hair shaft keratins and 51 keratin associated proteins, as well as desmosomal cadherins and several actors of the BMP signaling pathway which is important for hair shaft differentiation. The lower expression of keratin 71 and desmocollin 2 on the mRNA level in skin biopsies corresponded with a decreased protein expression in the hair shafts of affected dogs. The genetic analysis revealed a missense variant in the IGFBP5 gene homozygous in all available Greyhounds and other sighthounds. Further research is required to clarify whether the IGFBP5 variant represents a predisposing genetic risk factor. We conclude from our results that structural defects in the hair shafts are the cause for this well-known disease and these defects are associated with a downregulation of genes and proteins essential for hair shaft formation. Our data add important knowledge to further understand the molecular mechanisms of HF morphogenesis and alopecia in dogs.


Subject(s)
Alopecia , Dog Diseases , Hair , Skin , Alopecia/genetics , Alopecia/metabolism , Alopecia/pathology , Alopecia/veterinary , Animals , Dog Diseases/genetics , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Female , Gene Expression Regulation , Hair/metabolism , Hair/pathology , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Insulin-Like Growth Factor Binding Protein 5/genetics , Keratins/biosynthesis , Keratins/genetics , Male , Skin/metabolism , Skin/pathology
6.
Vet Dermatol ; 30(1): 64-e18, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30474267

ABSTRACT

BACKGROUND: The feline counterpart of human inflammatory linear verrucous epidermal nevus (ILVEN) has been described; however, the possible underlying developmental defect has not been investigated. OBJECTIVE: To report a case of multiple ILVEN-like lesions in a cat with a genetic variant in the NSDHL gene. ANIMALS: A 2-year-old, female, domestic short hair cat with a history of multiple alopecic, verrucous, hyperpigmented and erythematous skin lesions, following Blaschko's lines on the head, the limbs, the trunk and paw pads. METHODS AND RESULTS: According to the clinical and histopathological findings, a diagnosis of multiple ILVEN-like lesions was made. Genetic investigation revealed a heterozygous missense variant in the X-chromosomal NSDHL gene predicted to lead to a loss-of-function of the NSDHL protein. CONCLUSIONS AND CLINICAL IMPORTANCE: To the best of the authors' knowledge, this is the first case of feline ILVEN-like lesions in which a genetic cause has been proposed. Future studies to establish a causal relationship between NSDHL variants and skin lesions might lead to pathogenesis-directed treatments.


Subject(s)
3-Hydroxysteroid Dehydrogenases/genetics , Cat Diseases/genetics , Nevus/veterinary , Skin Neoplasms/veterinary , Animals , Cat Diseases/congenital , Cat Diseases/pathology , Cats , Female , Genes/genetics , Heterozygote , Mutation, Missense/genetics , Nevus/congenital , Nevus/genetics , Nevus/pathology , Skin/pathology , Skin Neoplasms/congenital , Skin Neoplasms/genetics , Skin Neoplasms/pathology
7.
PLoS Genet ; 14(3): e1007264, 2018 03.
Article in English | MEDLINE | ID: mdl-29565995

ABSTRACT

Lethal acrodermatitis (LAD) is a genodermatosis with monogenic autosomal recessive inheritance in Bull Terriers and Miniature Bull Terriers. The LAD phenotype is characterized by poor growth, immune deficiency, and skin lesions, especially at the paws. Utilizing a combination of genome wide association study and haplotype analysis, we mapped the LAD locus to a critical interval of ~1.11 Mb on chromosome 14. Whole genome sequencing of an LAD affected dog revealed a splice region variant in the MKLN1 gene that was not present in 191 control genomes (chr14:5,731,405T>G or MKLN1:c.400+3A>C). This variant showed perfect association in a larger combined Bull Terrier/Miniature Bull Terrier cohort of 46 cases and 294 controls. The variant was absent from 462 genetically diverse control dogs of 62 other dog breeds. RT-PCR analysis of skin RNA from an affected and a control dog demonstrated skipping of exon 4 in the MKLN1 transcripts of the LAD affected dog, which leads to a shift in the MKLN1 reading frame. MKLN1 encodes the widely expressed intracellular protein muskelin 1, for which diverse functions in cell adhesion, morphology, spreading, and intracellular transport processes are discussed. While the pathogenesis of LAD remains unclear, our data facilitate genetic testing of Bull Terriers and Miniature Bull Terriers to prevent the unintentional production of LAD affected dogs. This study may provide a starting point to further clarify the elusive physiological role of muskelin 1 in vivo.


Subject(s)
Acrodermatitis/veterinary , Cell Adhesion Molecules/genetics , Dog Diseases/genetics , Genes, Lethal , Intracellular Signaling Peptides and Proteins/genetics , RNA Splicing , Acrodermatitis/genetics , Animals , Chromosome Mapping , Dogs , Exons , Genome-Wide Association Study , Haplotypes , Real-Time Polymerase Chain Reaction
8.
PLoS One ; 12(10): e0185944, 2017.
Article in English | MEDLINE | ID: mdl-28973042

ABSTRACT

We investigated a German Spitz family where the mating of a black male to a white female had yielded three puppies with an unexpected light brown coat color, lightly pigmented lips and noses, and blue eyes. Combined linkage and homozygosity analysis based on a fully penetrant monogenic autosomal recessive mode of inheritance identified a critical interval of 15 Mb on chromosome 3. We obtained whole genome sequence data from one affected dog, three wolves, and 188 control dogs. Filtering for private variants revealed a single variant with predicted high impact in the critical interval in LOC100855460 (XM_005618224.1:c.377+2T>G LT844587.1:c.-45+2T>G). The variant perfectly co-segregated with the phenotype in the family. We genotyped 181 control dogs with normal pigmentation from diverse breeds including 22 unrelated German Spitz dogs, which were all homozygous wildtype. Comparative sequence analyses revealed that LOC100855460 actually represents the 5'-end of the canine OCA2 gene. The CanFam 3.1 reference genome assembly is incorrect and separates the first two exons from the remaining exons of the OCA2 gene. We amplified a canine OCA2 cDNA fragment by RT-PCR and determined the correct full-length mRNA sequence (LT844587.1). Variants in the OCA2 gene cause oculocutaneous albinism type 2 (OCA2) in humans, pink-eyed dilution in mice, and similar phenotypes in corn snakes, medaka and Mexican cave tetra fish. We therefore conclude that the observed oculocutaneous albinism in German Spitz is most likely caused by the identified variant in the 5'-splice site of the first intron of the canine OCA2 gene.


Subject(s)
Albinism, Oculocutaneous/veterinary , Dog Diseases/genetics , Dogs/genetics , Eye Color/genetics , Genotype , Membrane Transport Proteins/genetics , RNA Splicing , Albinism, Oculocutaneous/genetics , Animals , Exons , Female , Male , Pedigree
9.
PLoS One ; 12(8): e0180359, 2017.
Article in English | MEDLINE | ID: mdl-28846680

ABSTRACT

Keratins represent a large protein family with essential structural and functional roles in epithelial cells of skin, hair follicles, and other organs. During evolution the genes encoding keratins have undergone multiple rounds of duplication and humans have two clusters with a total of 55 functional keratin genes in their genomes. Due to the high similarity between different keratin paralogs and species-specific differences in gene content, the currently available keratin gene annotation in species with draft genome assemblies such as dog and horse is still imperfect. We compared the National Center for Biotechnology Information (NCBI) (dog annotation release 103, horse annotation release 101) and Ensembl (release 87) gene predictions for the canine and equine keratin gene clusters to RNA-seq data that were generated from adult skin of five dogs and two horses and from adult hair follicle tissue of one dog. Taking into consideration the knowledge on the conserved exon/intron structure of keratin genes, we annotated 61 putatively functional keratin genes in both the dog and horse, respectively. Subsequently, curators in the RefSeq group at NCBI reviewed their annotation of keratin genes in the dog and horse genomes (Annotation Release 104 and Annotation Release 102, respectively) and updated annotation and gene nomenclature of several keratin genes. The updates are now available in the NCBI Gene database (https://www.ncbi.nlm.nih.gov/gene).


Subject(s)
Databases, Genetic , Dogs/genetics , Genome , Horses/genetics , Keratins/genetics , Animals , Exons , Introns , Molecular Sequence Annotation , Species Specificity
10.
G3 (Bethesda) ; 7(9): 3115-3121, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28739597

ABSTRACT

In heterozygous females affected by an X-linked skin disorder, lesions often appear in a characteristic pattern, the so-called Blaschko's lines. We investigated a female Labrador Retriever and her crossbred daughter, which both showed similar clinical lesions that followed Blaschko's lines. The two male littermates of the affected daughter had died at birth, suggesting a monogenic X-chromosomal semidominant mode of inheritance. Whole genome sequencing of the affected daughter, and subsequent automated variant filtering with respect to 188 nonaffected control dogs of different breeds, revealed 332 hetero-zygous variants on the X-chromosome private to the affected dog. None of these variants was protein-changing. By visual inspection of candidate genes located on the X-chromosome, we identified a large deletion in the NSDHL gene, encoding NAD(P) dependent steroid dehydrogenase-like, a 3ß-hydroxysteroid dehydrogenase involved in cholesterol biosynthesis. The deletion spanned >14 kb, and included the last three exons of the NSDHL gene. By PCR and fragment length analysis, we confirmed the presence of the variant in both affected dogs, and its absence in 50 control Labrador Retrievers. Variants in the NSDHL gene cause CHILD syndrome in humans, and the bare patches (Bpa) and striated (Str) phenotypes in mice. Taken together, our genetic data and the known role of NSDHL in X-linked skin disorders strongly suggest that the identified structural variant in the NSDHL gene is causative for the phenotype in the two affected dogs.


Subject(s)
3-Hydroxysteroid Dehydrogenases/genetics , Congenital Abnormalities/veterinary , Dog Diseases/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Sequence Deletion , Animals , Biopsy , Dog Diseases/diagnosis , Dogs , Female , Genotype , Histocytochemistry , Phenotype , Skin/pathology , Whole Genome Sequencing
11.
PLoS Genet ; 13(3): e1006651, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28249031

ABSTRACT

Ichthyoses are a heterogeneous group of inherited cornification disorders characterized by generalized dry skin, scaling and/or hyperkeratosis. Ichthyosis vulgaris is the most common form of ichthyosis in humans and caused by genetic variants in the FLG gene encoding filaggrin. Filaggrin is a key player in the formation of the stratum corneum, the uppermost layer of the epidermis and therefore crucial for barrier function. During terminal differentiation of keratinocytes, the precursor profilaggrin is cleaved by several proteases into filaggrin monomers and eventually processed into free amino acids contributing to the hydration of the cornified layer. We studied a German Shepherd dog with a novel form of ichthyosis. Comparing the genome sequence of the affected dog with 288 genomes from genetically diverse non-affected dogs we identified a private heterozygous variant in the ASPRV1 gene encoding "aspartic peptidase, retroviral-like 1", which is also known as skin aspartic protease (SASPase). The variant was absent in both parents and therefore due to a de novo mutation event. It was a missense variant, c.1052T>C, affecting a conserved residue close to an autoprocessing cleavage site, p.(Leu351Pro). ASPRV1 encodes a retroviral-like protease involved in profilaggrin-to-filaggrin processing. By immunofluorescence staining we showed that the filaggrin expression pattern was altered in the affected dog. Thus, our findings provide strong evidence that the identified de novo variant is causative for the ichthyosis in the affected dog and that ASPRV1 plays an essential role in skin barrier formation. ASPRV1 is thus a novel candidate gene for unexplained human forms of ichthyoses.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Dog Diseases/genetics , Genetic Predisposition to Disease/genetics , Ichthyosis/genetics , Mutation, Missense , Amino Acid Sequence , Animals , Aspartic Acid Endopeptidases/metabolism , Base Sequence , Disease Models, Animal , Dog Diseases/enzymology , Dogs , Female , Filaggrin Proteins , Humans , Ichthyosis/enzymology , Ichthyosis/veterinary , Intermediate Filament Proteins/metabolism , Microscopy, Fluorescence , Sequence Analysis, DNA/methods , Sequence Homology, Amino Acid , Skin/enzymology , Skin/metabolism , Skin/pathology
12.
G3 (Bethesda) ; 7(4): 1315-1321, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28235824

ABSTRACT

Naked foal syndrome (NFS) is a genodermatosis in the Akhal-Teke horse breed. We provide the first scientific description of this phenotype. Affected horses have almost no hair and show a mild ichthyosis. So far, all known NFS affected horses died between a few weeks and 3 yr of age. It is not clear whether a specific pathology caused the premature deaths. NFS is inherited as a monogenic autosomal recessive trait. We mapped the disease causing genetic variant to two segments on chromosomes 7 and 27 in the equine genome. Whole genome sequencing of two affected horses, two obligate carriers, and 75 control horses from other breeds revealed a single nonsynonymous genetic variant on the chromosome 7 segment that was perfectly associated with NFS. The affected horses were homozygous for ST14:c.388G>T, a nonsense variant that truncates >80% of the open reading frame of the ST14 gene (p.Glu130*). The variant leads to partial nonsense-mediated decay of the mutant transcript. Genetic variants in the ST14 gene are responsible for autosomal recessive congenital ichthyosis 11 in humans. Thus, the identified equine ST14:c.388G>T variant is an excellent candidate causative variant for NFS, and the affected horses represent a large animal model for a known human genodermatosis. Our findings will enable genetic testing to avoid the nonintentional breeding of NFS-affected foals.


Subject(s)
Codon, Nonsense/genetics , Horse Diseases/genetics , Horses/genetics , Tumor Suppressor Proteins/genetics , Animals , Base Sequence , Chromosome Mapping , Female , Genetic Linkage , Genetic Predisposition to Disease , Heterozygote , Horse Diseases/pathology , Male , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Syndrome , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...