Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Vet Comp Oncol ; 21(4): 623-633, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37734854

RESUMO

Canine hemangiosarcoma (HSA) is an aggressive cancer of endothelial cells with short survival times. Understanding the genomic landscape of HSA may aid in developing therapeutic strategies for dogs and may also inform therapies for the rare and aggressive human cancer angiosarcoma. The objectives of this study were to build a framework for leveraging real-world genomic and clinical data that could provide the foundation for precision medicine in veterinary oncology, and to determine the relationships between genomic and clinical features in canine splenic HSA. One hundred and nine dogs with primary splenic HSA treated by splenectomy that had tumour sequencing via the FidoCure® Precision Medicine Platform targeted sequencing panel were enrolled. Patient signalment, weight, metastasis at diagnosis and overall survival time were retrospectively evaluated. The incidence of genomic alterations in individual genes and their relationship to patient variables including outcome were assessed. Somatic mutations in TP53 (n = 44), NRAS (n = 20) and PIK3CA (n = 19) were most common. Survival was associated with presence of metastases at diagnosis and germline variants in SETD2 and NOTCH1. Age at diagnosis was associated with somatic NRAS mutations and breed. TP53 and PIK3CA somatic mutations were found in larger dogs, while germline SETD2 variants were found in smaller dogs. We identified both somatic mutations and germline variants associated with clinical variables including age, breed and overall survival. These genetic changes may be useful prognostic factors and provide insight into the genomic landscape of hemangiosarcoma.


Assuntos
Doenças do Cão , Hemangiossarcoma , Neoplasias Esplênicas , Humanos , Cães , Animais , Hemangiossarcoma/genética , Hemangiossarcoma/veterinária , Hemangiossarcoma/tratamento farmacológico , Células Endoteliais , Estudos Retrospectivos , Doenças do Cão/genética , Doenças do Cão/tratamento farmacológico , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/veterinária , Neoplasias Esplênicas/tratamento farmacológico , Genômica , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/uso terapêutico
2.
Sci Rep ; 13(1): 10935, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37414794

RESUMO

Naturally occurring canine cancers have remarkable similarities to their human counterparts. To better understand these similarities, we investigated 671 client-owned dogs from 96 breeds with 23 common tumor types, including those whose mutation profile are unknown (anal sac carcinoma and neuroendocrine carcinoma) or understudied (thyroid carcinoma, soft tissue sarcoma and hepatocellular carcinoma). We discovered mutations in 50 well-established oncogenes and tumor suppressors, and compared them to those reported in human cancers. As in human cancer, TP53 is the most commonly mutated gene, detected in 22.5% of canine tumors overall. Canine tumors share mutational hotspots with human tumors in oncogenes including PIK3CA, KRAS, NRAS, BRAF, KIT and EGFR. Hotspot mutations with significant association to tumor type include NRAS G61R and PIK3CA H1047R in hemangiosarcoma, ERBB2 V659E in pulmonary carcinoma, and BRAF V588E (equivalent of V600E in humans) in urothelial carcinoma. Our findings better position canines as a translational model of human cancer to investigate a wide spectrum of targeted therapies.


Assuntos
Mutação , Neoplasias , Animais , Cães , Proteínas Oncogênicas/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Humanos , Antineoplásicos/uso terapêutico
3.
PLoS One ; 17(9): e0274383, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36099278

RESUMO

The characterization of immortalized canine osteosarcoma (OS) cell lines used for research has historically been based on phenotypic features such as cellular morphology and expression of bone specific markers. With the increasing use of these cell lines to investigate novel therapeutic approaches prior to in vivo translation, a much more detailed understanding regarding the genomic landscape of these lines is required to ensure accurate interpretation of findings. Here we report the first whole genome characterization of eight canine OS cell lines, including single nucleotide variants, copy number variants and other structural variants. Many alterations previously characterized in primary canine OS tissue were observed in these cell lines, including TP53 mutations, MYC copy number gains, loss of CDKN2A, PTEN, DLG2, MAGI2, and RB1 and structural variants involving SETD2, DLG2 and DMD. These data provide a new framework for understanding how best to incorporate in vitro findings generated using these cell lines into the design of future clinical studies involving dogs with spontaneous OS.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/veterinária , Linhagem Celular , Variações do Número de Cópias de DNA , Cães , Genômica , Osteossarcoma/genética , Osteossarcoma/veterinária
4.
Vet Comp Oncol ; 20(4): 797-804, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35574975

RESUMO

Although cancer is widely regarded as a major contributor to canine morbidity and mortality, its frequency in companion dogs has only infrequently been characterised. We analysed cross-sectional data from the baseline survey of owners of 27 541 living companion dogs enrolled in the Dog Aging Project as of 31 December 2020 to estimate the lifetime prevalence of malignant and benign tumours and several potentially-associated characteristics. Survey questions elicited information on history of 'cancer or tumors' including organ site and histologic type. Owners reported 819 malignant tumours (56% sited in the skin, muscle or other soft tissue) and 404 benign tumours (69% sited in the skin, muscle or other soft tissue). The lifetime prevalence of malignant tumours (29.7/1000 dogs) was approximately double the lifetime prevalence of benign tumours (14.7/1000 dogs). Lifetime prevalence of both malignant and benign tumours increased with dog age at survey completion. There were no statistically discernable differences in age-adjusted lifetime prevalence of malignant (prevalence ratio (PR) = 0.93 [95% confidence interval (CI) 0.82, 1.07] or benign tumours (PR = 1.10, 95% CI 0.91, 1.34) in mixed vs. purebred dogs. The lifetime prevalence of malignant tumours increased with increasing dog size class; compared to toy and small dogs, the age-adjusted PRs (95% CIs) for medium, standard, large, and giant dogs were 1.65 (1.28, 2.11), 2.92 (2.35, 3.64), 3.67 (2.92, 4.62) and 2.99 (1.23, 4.02), respectively. Similar though less pronounced patterns in relation to dog size class were observed for benign tumours. Ongoing prospective data collection will permit future studies on risk factors for canine tumour incidence.


Assuntos
Doenças do Cão , Neoplasias , Cães , Animais , Estudos Transversais , Animais de Estimação , Prevalência , Doenças do Cão/epidemiologia , Doenças do Cão/patologia , Neoplasias/epidemiologia , Neoplasias/veterinária , Neoplasias/patologia , Envelhecimento , Inquéritos e Questionários
5.
Science ; 376(6592): eabk0639, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35482869

RESUMO

Behavioral genetics in dogs has focused on modern breeds, which are isolated subgroups with distinctive physical and, purportedly, behavioral characteristics. We interrogated breed stereotypes by surveying owners of 18,385 purebred and mixed-breed dogs and genotyping 2155 dogs. Most behavioral traits are heritable [heritability (h2) > 25%], and admixture patterns in mixed-breed dogs reveal breed propensities. Breed explains just 9% of behavioral variation in individuals. Genome-wide association analyses identify 11 loci that are significantly associated with behavior, and characteristic breed behaviors exhibit genetic complexity. Behavioral loci are not unusually differentiated in breeds, but breed propensities align, albeit weakly, with ancestral function. We propose that behaviors perceived as characteristic of modern breeds derive from thousands of years of polygenic adaptation that predates breed formation, with modern breeds distinguished primarily by aesthetic traits.


Assuntos
Estudo de Associação Genômica Ampla , Genômica , Animais , Cruzamento , Cães , Fenótipo
6.
Genes (Basel) ; 12(12)2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34946912

RESUMO

Dogs represent a unique spontaneous cancer model. Osteosarcoma (OSA) is the most common primary bone tumor in dogs (OMIA 001441-9615), and strongly resembles human forms of OSA. Several large- to giant-sized dog breeds, including the Leonberger, have a greatly increased risk of developing OSA. We performed genome-wide association analysis with high-density imputed SNP genotype data from 273 Leonberger cases with a median age of 8.1 [3.1-13.5] years and 365 controls older than eight years. This analysis revealed significant associations at the CDKN2A/B gene locus on canine chromosome 11, mirroring previous findings in other dog breeds, such as the greyhound, that also show an elevated risk for OSA. Heritability (h2SNP) was determined to be 20.6% (SE = 0.08; p-value = 5.7 × 10-4) based on a breed prevalence of 20%. The 2563 SNPs across the genome accounted for nearly all the h2SNP of OSA, with 2183 SNPs of small effect, 316 SNPs of moderate effect, and 64 SNPs of large effect. As with many other cancers it is likely that regulatory, non-coding variants underlie the increased risk for cancer development. Our findings confirm a complex genetic basis of OSA, moderate heritability, and the crucial role of the CDKN2A/B locus leading to strong cancer predisposition in dogs. It will ultimately be interesting to study and compare the known genetic loci associated with canine OSA in human OSA.


Assuntos
Neoplasias Ósseas/patologia , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Doenças do Cão/patologia , Loci Gênicos , Osteossarcoma/patologia , Polimorfismo de Nucleotídeo Único , Animais , Neoplasias Ósseas/genética , Doenças do Cão/genética , Cães , Predisposição Genética para Doença , Genoma , Estudo de Associação Genômica Ampla , Osteossarcoma/genética
7.
Mol Cancer Res ; 19(5): 847-861, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33649193

RESUMO

Sporadic angiosarcomas are aggressive vascular sarcomas whose rarity and genomic complexity present significant obstacles in deciphering the pathogenic significance of individual genetic alterations. Numerous fusion genes have been identified across multiple types of cancers, but their existence and significance remain unclear in sporadic angiosarcomas. In this study, we leveraged RNA-sequencing data from 13 human angiosarcomas and 76 spontaneous canine hemangiosarcomas to identify fusion genes associated with spontaneous vascular malignancies. Ten novel protein-coding fusion genes, including TEX2-PECAM1 and ATP8A2-FLT1, were identified in seven of the 13 human tumors, with two tumors showing mutations of TP53. HRAS and NRAS mutations were found in angiosarcomas without fusions or TP53 mutations. We found 15 novel protein-coding fusion genes including MYO16-PTK2, GABRA3-FLT1, and AKT3-XPNPEP1 in 11 of the 76 canine hemangiosarcomas; these fusion genes were seen exclusively in tumors of the angiogenic molecular subtype that contained recurrent mutations in TP53, PIK3CA, PIK3R1, and NRAS. In particular, fusion genes and mutations of TP53 cooccurred in tumors with higher frequency than expected by random chance, and they enriched gene signatures predicting activation of angiogenic pathways. Comparative transcriptomic analysis of human angiosarcomas and canine hemangiosarcomas identified shared molecular signatures associated with activation of PI3K/AKT/mTOR pathways. Our data suggest that genome instability induced by TP53 mutations might create a predisposition for fusion events that may contribute to tumor progression by promoting selection and/or enhancing fitness through activation of convergent angiogenic pathways in this vascular malignancy. IMPLICATIONS: This study shows that, while drive events of malignant vasoformative tumors of humans and dogs include diverse mutations and stochastic rearrangements that create novel fusion genes, convergent transcriptional programs govern the highly conserved morphologic organization and biological behavior of these tumors in both species.


Assuntos
Doenças do Cão/genética , Perfilação da Expressão Gênica/métodos , Hemangiossarcoma/genética , Neoplasias Vasculares/genética , Animais , Cães , Fusão Gênica , Genômica/métodos , Humanos , Transcrição Gênica
8.
Mol Biol Evol ; 37(1): 11-17, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31688937

RESUMO

Despite a considerable expenditure of time and resources and significant advances in experimental models of disease, cancer research continues to suffer from extremely low success rates in translating preclinical discoveries into clinical practice. The continued failure of cancer drug development, particularly late in the course of human testing, not only impacts patient outcomes, but also drives up the cost for those therapies that do succeed. It is clear that a paradigm shift is necessary if improvements in this process are to occur. One promising direction for increasing translational success is comparative oncology-the study of cancer across species, often involving veterinary patients that develop naturally-occurring cancers. Comparative oncology leverages the power of cross-species analyses to understand the fundamental drivers of cancer protective mechanisms, as well as factors contributing to cancer initiation and progression. Clinical trials in veterinary patients with cancer provide an opportunity to evaluate novel therapeutics in a setting that recapitulates many of the key features of human cancers, including genomic aberrations that underly tumor development, response and resistance to treatment, and the presence of comorbidities that can affect outcomes. With a concerted effort from basic scientists, human physicians and veterinarians, comparative oncology has the potential to enhance the cost-effectiveness and efficiency of pipelines for cancer drug discovery and other cancer treatments.


Assuntos
Descoberta de Drogas , Neoplasias/veterinária , Animais , Humanos , Neoplasias/tratamento farmacológico
9.
Mol Biol Evol ; 37(2): 320-326, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31642480

RESUMO

Cancer progression is an evolutionary process. During this process, evolving cancer cell populations encounter restrictive ecological niches within the body, such as the primary tumor, circulatory system, and diverse metastatic sites. Efforts to prevent or delay cancer evolution-and progression-require a deep understanding of the underlying molecular evolutionary processes. Herein we discuss a suite of concepts and tools from evolutionary and ecological theory that can inform cancer biology in new and meaningful ways. We also highlight current challenges to applying these concepts, and propose ways in which incorporating these concepts could identify new therapeutic modes and vulnerabilities in cancer.


Assuntos
Genômica/métodos , Neoplasias/genética , Progressão da Doença , Evolução Molecular , Aptidão Genética , Humanos , Filogenia , Nicho de Células-Tronco
10.
Mol Cancer Res ; 17(12): 2410-2421, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31570656

RESUMO

Angiosarcoma is a highly aggressive cancer of blood vessel-forming cells with few effective treatment options and high patient mortality. It is both rare and heterogenous, making large, well-powered genomic studies nearly impossible. Dogs commonly suffer from a similar cancer, called hemangiosarcoma, with breeds like the golden retriever carrying heritable genetic factors that put them at high risk. If the clinical similarity of canine hemangiosarcoma and human angiosarcoma reflects shared genomic etiology, dogs could be a critically needed model for advancing angiosarcoma research. We assessed the genomic landscape of canine hemangiosarcoma via whole-exome sequencing (47 golden retriever hemangiosarcomas) and RNA sequencing (74 hemangiosarcomas from multiple breeds). Somatic coding mutations occurred most frequently in the tumor suppressor TP53 (59.6% of cases) as well as two genes in the PI3K pathway: the oncogene PIK3CA (29.8%) and its regulatory subunit PIK3R1 (8.5%). The predominant mutational signature was the age-associated deamination of cytosine to thymine. As reported in human angiosarcoma, CDKN2A/B was recurrently deleted and VEGFA, KDR, and KIT recurrently gained. We compared the canine data to human data recently released by The Angiosarcoma Project, and found many of the same genes and pathways significantly enriched for somatic mutations, particularly in breast and visceral angiosarcomas. Canine hemangiosarcoma closely models the genomic landscape of human angiosarcoma of the breast and viscera, and is a powerful tool for investigating the pathogenesis of this devastating disease. IMPLICATIONS: We characterize the genomic landscape of canine hemangiosarcoma and demonstrate its similarity to human angiosarcoma.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Hemangiossarcoma/genética , Proteína Supressora de Tumor p53/genética , Animais , Vasos Sanguíneos/patologia , Mama/metabolismo , Mama/patologia , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Cães , Feminino , Genoma/genética , Genômica , Hemangiossarcoma/patologia , Humanos , Mutação/genética , Vísceras/metabolismo , Vísceras/patologia , Sequenciamento do Exoma
11.
Genes (Basel) ; 10(6)2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31181663

RESUMO

Dogs are an unparalleled natural model for investigating the genetics of health and disease, particularly for complex diseases like cancer. Comprehensive genomic annotation of regulatory elements active in healthy canine tissues is crucial both for identifying candidate causal variants and for designing functional studies needed to translate genetic associations into disease insight. Currently, canine geneticists rely primarily on annotations of the human or mouse genome that have been remapped to dog, an approach that misses dog-specific features. Here, we describe BarkBase, a canine epigenomic resource available at barkbase.org. BarkBase hosts data for 27 adult tissue types, with biological replicates, and for one sample of up to five tissues sampled at each of four carefully staged embryonic time points. RNA sequencing is complemented with whole genome sequencing and with assay for transposase-accessible chromatin using sequencing (ATAC-seq), which identifies open chromatin regions. By including replicates, we can more confidently discern tissue-specific transcripts and assess differential gene expression between tissues and timepoints. By offering data in easy-to-use file formats, through a visual browser modeled on similar genomic resources for human, BarkBase introduces a powerful new resource to support comparative studies in dogs and humans.


Assuntos
Cromatina/genética , Epigenômica , Genoma/genética , Análise de Sequência de DNA , Adulto , Animais , Cães , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Anotação de Sequência Molecular , Sequências Reguladoras de Ácido Nucleico/genética , Análise de Sequência de RNA , Software
12.
Proc Natl Acad Sci U S A ; 113(22): E3091-100, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27185954

RESUMO

Canine degenerative myelopathy (DM) is a naturally occurring neurodegenerative disease with similarities to some forms of amyotrophic lateral sclerosis (ALS). Most dogs that develop DM are homozygous for a common superoxide dismutase 1 gene (SOD1) mutation. However, not all dogs homozygous for this mutation develop disease. We performed a genome-wide association analysis in the Pembroke Welsh Corgi (PWC) breed comparing DM-affected and -unaffected dogs homozygous for the SOD1 mutation. The analysis revealed a modifier locus on canine chromosome 25. A haplotype within the SP110 nuclear body protein (SP110) was present in 40% of affected compared with 4% of unaffected dogs (P = 1.5 × 10(-5)), and was associated with increased probability of developing DM (P = 4.8 × 10(-6)) and earlier onset of disease (P = 1.7 × 10(-5)). SP110 is a nuclear body protein involved in the regulation of gene transcription. Our findings suggest that variations in SP110-mediated gene transcription may underlie, at least in part, the variability in risk for developing DM among PWCs that are homozygous for the disease-related SOD1 mutation. Further studies are warranted to clarify the effect of this modifier across dog breeds.


Assuntos
Doenças do Cão/genética , Doenças Musculares/genética , Mutação/genética , Doenças Neurodegenerativas/genética , Proteínas Nucleares/genética , Doenças da Medula Espinal/genética , Superóxido Dismutase/genética , Idade de Início , Animais , Modelos Animais de Doenças , Doenças do Cão/patologia , Cães , Feminino , Estudo de Associação Genômica Ampla , Homozigoto , Masculino , Doenças Musculares/patologia , Doenças Neurodegenerativas/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doenças da Medula Espinal/patologia
13.
PLoS Genet ; 11(2): e1004922, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25642983

RESUMO

Dogs, with their breed-determined limited genetic background, are great models of human disease including cancer. Canine B-cell lymphoma and hemangiosarcoma are both malignancies of the hematologic system that are clinically and histologically similar to human B-cell non-Hodgkin lymphoma and angiosarcoma, respectively. Golden retrievers in the US show significantly elevated lifetime risk for both B-cell lymphoma (6%) and hemangiosarcoma (20%). We conducted genome-wide association studies for hemangiosarcoma and B-cell lymphoma, identifying two shared predisposing loci. The two associated loci are located on chromosome 5, and together contribute ~20% of the risk of developing these cancers. Genome-wide p-values for the top SNP of each locus are 4.6×10-7 and 2.7×10-6, respectively. Whole genome resequencing of nine cases and controls followed by genotyping and detailed analysis identified three shared and one B-cell lymphoma specific risk haplotypes within the two loci, but no coding changes were associated with the risk haplotypes. Gene expression analysis of B-cell lymphoma tumors revealed that carrying the risk haplotypes at the first locus is associated with down-regulation of several nearby genes including the proximal gene TRPC6, a transient receptor Ca2+-channel involved in T-cell activation, among other functions. The shared risk haplotype in the second locus overlaps the vesicle transport and release gene STX8. Carrying the shared risk haplotype is associated with gene expression changes of 100 genes enriched for pathways involved in immune cell activation. Thus, the predisposing germ-line mutations in B-cell lymphoma and hemangiosarcoma appear to be regulatory, and affect pathways involved in T-cell mediated immune response in the tumor. This suggests that the interaction between the immune system and malignant cells plays a common role in the tumorigenesis of these relatively different cancers.


Assuntos
Carcinogênese/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Hemangiossarcoma/genética , Linfoma de Células B/genética , Animais , Linfócitos B/patologia , Cruzamento , Carcinogênese/imunologia , Cães , Genótipo , Mutação em Linhagem Germinativa , Haplótipos/genética , Hemangiossarcoma/imunologia , Hemangiossarcoma/patologia , Hemangiossarcoma/veterinária , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Linfoma de Células B/veterinária , Polimorfismo de Nucleotídeo Único , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA