Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Vet Anaesth Analg ; 50(4): 341-348, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37291042

RESUMO

OBJECTIVE: To investigate the statistical association of severe intraoperative hypoxemia in thoracic surgery with mortality, postoperative hospitalization times and cost of care. STUDY DESIGN: Retrospective study. ANIMALS: Dogs that underwent thoracic surgery in three veterinary hospitals between October 1, 2018 and October 1, 2020. METHODS: Anesthesia and hospitalization records from 112 dogs were reviewed and 94 cases met inclusion criteria. Recorded data included signalment, disease etiology, pulmonary or extrapulmonary nature of disease, surgical procedure performed, episodes of severe intraoperative hypoxemia defined as a pulse oximetry reading (SpO2) <90% of 5 minutes or longer duration, survival to discharge, time from extubation to hospital discharge and total invoice cost for clinical visit. Dogs were divided into two groups, those that experienced severe hypoxemia (group A) and those in which SpO2 reading <90% was not observed throughout the procedure (group B). RESULTS: Group A had a greater risk of mortality (odds ratio 10.6, 95% confidence interval 1.9-106.7; p = 0.002), prolonged hospitalization (median 62 hours versus 46 hours; p = 0.035) and more expensive cost of care (median US$10,287 versus $8506; p = 0.056) than group B. No significant difference was found for the type of surgical procedure or pulmonary versus extrapulmonary nature of disease. CONCLUSIONS AND CLINICAL RELEVANCE: Severe intraoperative hypoxemia was statistically associated with an increased risk of mortality and longer postoperative hospitalization times. Although not achieving statistical significance, there was a trend toward increased costs to the client for animals with intraoperative hypoxemia.


Assuntos
Cirurgia Torácica , Procedimentos Cirúrgicos Torácicos , Cães , Animais , Estudos Retrospectivos , Hipóxia/etiologia , Hipóxia/veterinária , Procedimentos Cirúrgicos Torácicos/efeitos adversos , Procedimentos Cirúrgicos Torácicos/veterinária , Oximetria/veterinária
2.
PLoS Genet ; 14(9): e1007589, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30188888

RESUMO

Canine malignant melanoma, a significant cause of mortality in domestic dogs, is a powerful comparative model for human melanoma, but little is known about its genetic etiology. We mapped the genomic landscape of canine melanoma through multi-platform analysis of 37 tumors (31 mucosal, 3 acral, 2 cutaneous, and 1 uveal) and 17 matching constitutional samples including long- and short-insert whole genome sequencing, RNA sequencing, array comparative genomic hybridization, single nucleotide polymorphism array, and targeted Sanger sequencing analyses. We identified novel predominantly truncating mutations in the putative tumor suppressor gene PTPRJ in 19% of cases. No BRAF mutations were detected, but activating RAS mutations (24% of cases) occurred in conserved hotspots in all cutaneous and acral and 13% of mucosal subtypes. MDM2 amplifications (24%) and TP53 mutations (19%) were mutually exclusive. Additional low-frequency recurrent alterations were observed amidst low point mutation rates, an absence of ultraviolet light mutational signatures, and an abundance of copy number and structural alterations. Mutations that modulate cell proliferation and cell cycle control were common and highlight therapeutic axes such as MEK and MDM2 inhibition. This mutational landscape resembles that seen in BRAF wild-type and sun-shielded human melanoma subtypes. Overall, these data inform biological comparisons between canine and human melanoma while suggesting actionable targets in both species.


Assuntos
Melanoma/genética , Melanoma/veterinária , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/veterinária , Animais , Ciclo Celular/genética , Proliferação de Células/genética , Hibridização Genômica Comparativa , Análise Mutacional de DNA , Doenças do Cão/genética , Cães , Feminino , Masculino , Melanoma/sangue , Melanoma/patologia , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais/genética , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/patologia , Análise Serial de Tecidos
3.
Cancer ; 125(20): 3514-3525, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31355930

RESUMO

Patients who are diagnosed with osteosarcoma (OS) today receive the same therapy that patients have received over the last 4 decades. Extensive efforts to identify more effective or less toxic regimens have proved disappointing. As we enter a postgenomic era in which we now recognize OS not as a cancer of mutations but as one defined by p53 loss, chromosomal complexity, copy number alteration, and profound heterogeneity, emerging threads of discovery leave many hopeful that an improving understanding of biology will drive discoveries that improve clinical care. Under the organization of the Bone Tumor Biology Committee of the Children's Oncology Group, a team of clinicians and scientists sought to define the state of the science and to identify questions that, if answered, have the greatest potential to drive fundamental clinical advances. Having discussed these questions in a series of meetings, each led by invited experts, we distilled these conversations into a series of seven Provocative Questions. These include questions about the molecular events that trigger oncogenesis, the genomic and epigenomic drivers of disease, the biology of lung metastasis, research models that best predict clinical outcomes, and processes for translating findings into clinical trials. Here, we briefly present each Provocative Question, review the current scientific evidence, note the immediate opportunities, and speculate on the impact that answered questions might have on the field. We do so with an intent to provide a framework around which investigators can build programs and collaborations to tackle the hardest problems and to establish research priorities for those developing policies and providing funding.


Assuntos
Epigenômica , Genômica , Osteossarcoma/terapia , Pesquisa Translacional Biomédica , Criança , Humanos , Mutação/genética , Osteossarcoma/epidemiologia , Osteossarcoma/genética , Osteossarcoma/patologia , Proteômica , Proteína Supressora de Tumor p53/genética
4.
PLoS Genet ; 12(2): e1005884, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26925584

RESUMO

The inactivation of p53 creates a major challenge for inducing apoptosis in cancer cells. An attractive strategy is to identify and subsequently target the survival signals in p53 defective cancer cells. Here we uncover a RUNX2-mediated survival signal in p53 defective cancer cells. The inhibition of this signal induces apoptosis in cancer cells but not non-transformed cells. Using the CRISPR technology, we demonstrate that p53 loss enhances the apoptosis caused by RUNX2 knockdown. Mechanistically, RUNX2 provides the survival signal partially through inducing MYC transcription. Cancer cells have high levels of activating histone marks on the MYC locus and concomitant high MYC expression. RUNX2 knockdown decreases the levels of these histone modifications and the recruitment of the Menin/MLL1 (mixed lineage leukemia 1) complex to the MYC locus. Two inhibitors of the Menin/MLL1 complex induce apoptosis in p53 defective cancer cells. Together, we identify a RUNX2-mediated epigenetic mechanism of the survival of p53 defective cancer cells and provide a proof-of-principle that the inhibition of this epigenetic axis is a promising strategy to kill p53 defective cancer cells.


Assuntos
Neoplasias Ósseas/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Epigênese Genética , Osteossarcoma/genética , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/genética , Sítios de Ligação , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes myc , Humanos , Camundongos Knockout , Osteossarcoma/patologia , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Proc Natl Acad Sci U S A ; 111(17): 6323-8, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24733928

RESUMO

Inflammation is widely recognized as an inducer of cancer progression. The inflammation-associated enzyme, inducible nitric oxide synthase (NOS2), has emerged as a candidate oncogene in estrogen receptor (ER)-negative breast cancer, and its increased expression is associated with disease aggressiveness and poor survival. Although these observations implicate NOS2 as an attractive therapeutic target, the mechanisms of both NOS2 induction in tumors and nitric oxide (NO)-driven cancer progression are not fully understood. To enhance our mechanistic understanding of NOS2 induction in tumors and its role in tumor biology, we used stimulants of NOS2 expression in ER(-) and ER(+) breast cancer cells and examined downstream NO-dependent effects. Herein, we show that up-regulation of NOS2 occurs in response to hypoxia, serum withdrawal, IFN-γ, and exogenous NO, consistent with a feed-forward regulation of NO production by the tumor microenvironment in breast cancer biology. Moreover, we found that key indicators of an aggressive cancer phenotype including increased S100 calcium binding protein A8, IL-6, IL-8, and tissue inhibitor matrix metalloproteinase-1 are up-regulated by these NOS2 stimulants, whereas inhibition of NOS2 in MDA-MB-231 breast cancer cells suppressed these markers. Moreover, NO altered cellular migration and chemoresistance of MDA-MB-231 cells to Taxol. Most notably, MDA-MB-231 tumor xenographs and cell metastases from the fat pad to the brain were significantly suppressed by NOS2 inhibition in nude mice. In summary, these results link elevated NOS2 to signals from the tumor microenvironment that arise with cancer progression and show that NO production regulates chemoresistance and metastasis of breast cancer cells.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Progressão da Doença , Retroalimentação Fisiológica , Óxido Nítrico Sintase Tipo II/metabolismo , Microambiente Tumoral , Animais , Biomarcadores Tumorais/metabolismo , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Meios de Cultura Livres de Soro , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Guanidinas , Humanos , Interferon gama/farmacologia , Camundongos , Modelos Biológicos , Metástase Neoplásica , Óxido Nítrico/farmacologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
6.
Am J Pathol ; 185(5): 1471-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25795282

RESUMO

Breast cancer metastasis is the leading cause of cancer-related deaths in women worldwide. Collagen in the tumor microenvironment plays a crucial role in regulating tumor progression. We have shown that type III collagen (Col3), a component of tumor stroma, regulates myofibroblast differentiation and scar formation after cutaneous injury. During the course of these wound-healing studies, we noted that tumors developed at a higher frequency in Col3(+/-) mice compared to wild-type littermate controls. We, therefore, examined the effect of Col3 deficiency on tumor behavior, using the murine mammary carcinoma cell line 4T1. Notably, tumor volume and pulmonary metastatic burden after orthotopic injection of 4T1 cells were increased in Col3(+/-) mice compared to Col3(+/+) littermates. By using murine (4T1) and human (MDA-MB-231) breast cancer cells grown in Col3-poor and Col3-enriched microenvironments in vitro, we found that several major events of the metastatic process were suppressed by Col3, including adhesion, invasion, and migration. In addition, Col3 deficiency increased proliferation and decreased apoptosis of 4T1 cells both in vitro and in primary tumors in vivo. Mechanistically, Col3 suppresses the procarcinogenic microenvironment by regulating stromal organization, including density and alignment of fibrillar collagen and myofibroblasts. We propose that Col3 plays an important role in the tumor microenvironment by suppressing metastasis-promoting characteristics of the tumor-associated stroma.


Assuntos
Colágeno Tipo III/metabolismo , Neoplasias Mamárias Experimentais/patologia , Invasividade Neoplásica/patologia , Microambiente Tumoral/fisiologia , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
7.
Stem Cells ; 33(4): 1304-19, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25524638

RESUMO

The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Proteína Supressora de Tumor p53/deficiência , Animais , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Camundongos , Camundongos Knockout
8.
Pediatr Blood Cancer ; 62(3): 450-5, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25611047

RESUMO

BACKGROUND: Survival rates of patients with osteosarcoma have remained stagnant over the last thirty years. Better understanding of biology, new therapeutics, and improved biomarkers are needed. The Children's Oncology Group (COG) addressed this need by developing one of the largest osteosarcoma biorepositories ever, containing over 15,000 tumor and tissue samples from over 1,500 patients. PROCEDURE: The biology study P9851 and the banking study AOST06B1 has enrolled 1,787 patients (as of September, 2013). Clinical information was lacking on 510 patients on P9851, who were not enrolled on a concurrent therapeutic trial. The value of these specimens was diminished. The lack of statistical support available for biology projects slowed the analysis of several critical studies. The QuadW Foundation, CureSearch, and the COG formed the Childhood Sarcoma Biostatistics and Annotation Office (CSBAO) to provide the infrastructure and address these needs by linking clinically annotated patient data to archived tissue samples and to develop biostatistical support for childhood sarcoma research. RESULTS: Originally 5.3% of samples from the 510 patients on P9851 not enrolled on a therapeutic study had full clinical annotation. The efforts of the CSBAO have linked clinical annotation to 90.8% of those specimens and provided statistical analyses to several studies that had used COG samples. As a result, 24 biology studies in osteosarcoma have been completed and published in peer-reviewed journals. CONCLUSIONS: These samples and in-silico data are available to the research community for basic and translational science projects to improve the biological understanding and treatment of patients affected by osteosarcoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Bases de Dados Factuais , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Bancos de Tecidos , Adolescente , Adulto , Criança , Pré-Escolar , Ensaios Clínicos Fase II como Assunto , Feminino , Humanos , Masculino , Ensaios Clínicos Controlados Aleatórios como Assunto
9.
Adv Exp Med Biol ; 804: 181-201, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24924175

RESUMO

The cause of death for the vast majority of cancer patients is the development of metastases at sites distant from that of the primary tumor. For most pediatric sarcoma patients such as those with osteosarcoma (OS), despite successful management of the primary tumor through multimodality approaches, the development of metastases, commonly to the lungs, is the cause of death. Significant improvements in long-term outcome for these patients have not been seen in more than 30 years. Furthermore, the long-term outcome for patients who present with metastatic disease is grave [1-5]. New treatment options are needed.Opportunities to improve outcomes for patients who present with metastases and those at-risk for progression and metastasis require an improved understanding of cancer progression and metastasis. With this goal in mind we and others have identified ezrin as a metastasis-associated protein that associated with OS and other cancers. Ezrin is the prototypical ERM (Ezrin/Radixin/Moesin) protein family member. ERMs function as linker proteins connecting the actin cytoskeleton and the plasma membrane. Since our initial identification of ezrin in pediatric sarcoma, an increasing understanding the role of ezrin in metastasis has emerged. Briefly, ezrin appears to allow metastatic cells to overcome a number of stresses experienced during the metastatic cascade, most notably the stress experienced as cells interact with the microenvironment of the secondary site. Cells must rapidly adapt to this environment in order to survive. Evidence now suggests a connection between ezrin expression and a variety of mechanisms linked to this important cellular adaptation including the ability of metastatic cells to initiate the translation of new proteins and to allow the efficient generation of ATP through a variety of sources. This understanding of the role of ezrin in the biology of metastasis is now sufficient to consider ezrin as an important therapeutic target in osteosarcoma patients. This chapter reviews our understanding of ezrin and the related ERM proteins in normal tissues and physiology, summarizes the expression of ezrin in human cancers and associations with clinical parameters of disease progression, reviews reports that detail a biological understanding of ezrin's role in metastatic progression, and concludes with a rationale that may be considered to target ezrin and ezrin biology in osteosarcoma.


Assuntos
Neoplasias Ósseas/genética , Proteínas do Citoesqueleto/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Osteossarcoma/genética , Adamantano/análogos & derivados , Adamantano/farmacologia , Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Criança , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Terapia de Alvo Molecular , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/secundário , Fenóis/farmacologia , Quinolinas/farmacologia , Quinolonas/farmacologia , Transdução de Sinais
10.
Am J Vet Res ; 85(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37913632

RESUMO

Necrotizing meningoencephalitis (NME) is a fatal neuroinflammatory disease that previously carried a uniformly grave prognosis. Our recent identification of a novel early form of NME in Pugs suggests that disease onset and progression are likely more insidious than previously recognized and provides new hope that early therapeutic intervention may halt disease progression and ultimately prevent or cure NME. This novel perspective also sheds new light on the clinical similarities to multiple sclerosis (MS) in humans and provides a rationale for cross-species translation. The history of recent scientific discoveries in NME and new parallels between MS and NME will be reviewed.


Assuntos
Doenças do Cão , Meningoencefalite , Esclerose Múltipla , Humanos , Cães , Animais , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/veterinária , Meningoencefalite/diagnóstico , Meningoencefalite/veterinária , Meningoencefalite/genética , Fenótipo , Doenças do Cão/genética
11.
iScience ; 26(2): 106070, 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36824269

RESUMO

PTEN encodes a tumor suppressor with lipid and protein phosphatase activities whose dysfunction has been implicated in melanomagenesis; less is known about how its phosphatases regulate melanoma metastasis. We demonstrate that PTEN expression negatively correlates with metastatic progression in human melanoma samples and a PTEN-deficient mouse melanoma model. Wildtype PTEN expression inhibited melanoma cell invasiveness and metastasis in a dose-dependent manner, behaviors that specifically required PTEN protein phosphatase activity. PTEN phosphatase activity regulated metastasis through Entpd5. Entpd5 knockdown reduced metastasis and IGF1R levels while promoting ER stress. In contrast, Entpd5 overexpression promoted metastasis and enhanced IGF1R levels while reducing ER stress. Moreover, Entpd5 expression was regulated by the ER stress sensor ATF6. Altogether, our data indicate that PTEN phosphatase activity inhibits metastasis by negatively regulating the Entpd5/IGF1R pathway through ATF6, thereby identifying novel candidate therapeutic targets for the treatment of PTEN mutant melanoma.

12.
JCO Clin Cancer Inform ; 7: e2300104, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37956387

RESUMO

PURPOSE: Osteosarcoma research advancement requires enhanced data integration across different modalities and sources. Current osteosarcoma research, encompassing clinical, genomic, protein, and tissue imaging data, is hindered by the siloed landscape of data generation and storage. MATERIALS AND METHODS: Clinical, molecular profiling, and tissue imaging data for 573 patients with pediatric osteosarcoma were collected from four public and institutional sources. A common data model incorporating standardized terminology was created to facilitate the transformation, integration, and load of source data into a relational database. On the basis of this database, a data commons accompanied by a user-friendly web portal was developed, enabling various data exploration and analytics functions. RESULTS: The Osteosarcoma Explorer (OSE) was released to the public in 2021. Leveraging a comprehensive and harmonized data set on the backend, the OSE offers a wide range of functions, including Cohort Discovery, Patient Dashboard, Image Visualization, and Online Analysis. Since its initial release, the OSE has experienced an increasing utilization by the osteosarcoma research community and provided solid, continuous user support. To our knowledge, the OSE is the largest (N = 573) and most comprehensive research data commons for pediatric osteosarcoma, a rare disease. This project demonstrates an effective framework for data integration and data commons development that can be readily applied to other projects sharing similar goals. CONCLUSION: The OSE offers an online exploration and analysis platform for integrated clinical, molecular profiling, and tissue imaging data of osteosarcoma. Its underlying data model, database, and web framework support continuous expansion onto new data modalities and sources.


Assuntos
Gerenciamento de Dados , Osteossarcoma , Criança , Humanos , Bases de Dados Factuais , Genômica , Osteossarcoma/diagnóstico por imagem , Osteossarcoma/genética
13.
J Vet Intern Med ; 36(4): 1365-1372, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35521894

RESUMO

BACKGROUND: Intervertebral disc-associated epidural hemorrhage (EH) in dogs is a poorly understood neurological condition. OBJECTIVE: To compare the clinical presentation, magnetic resonance imaging (MRI) changes, and clinical outcome of dogs with acute thoracolumbar intervertebral disc herniation (TL-IVDH) with and without EH. ANIMALS: One hundred sixty client-owned dogs that underwent MRI and hemilaminectomy for acute TL-IVDH at a private practice in Colorado, including 63 dogs with EH and 97 dogs without EH. METHODS: Retrospective review of medical record data from 160 dogs presenting sequentially to a single practice with acute TL-IVDH that underwent MRI and hemilaminectomy surgery. RESULTS: Sixty-three of 160 (39%) dogs had confirmed EH. French Bulldogs were significantly overrepresented (23/63; odds ratio [OR]: 4.1; 95% confidence interval [CI]: 1.8-9.0; P < .001) of the EH cases. Dogs with EH were more likely to present with clinical signs less than 48 hours than were dogs without EH (24-48 vs 48-72 hours; OR: 2.4; 95% CI: 1.2-4.6; P = .02) and were more likely to be nonambulatory on presentation (OR: 2.1; 95% CI: 1.0-4.1; P = .04). Dogs with EH were more likely to have <50% cross-sectional spinal cord compression than dogs without EH (OR: 2.3 vs. 0.4; 95% CI: 1.2-4.4 and 0.2-0.9, respectively), longer longitudinal spinal cord compression (3 spaces vs 1 space, P < .001), and greater intrinsic spinal cord change (grade 3/severe vs grade 1/mild; P < .001) based on MRI. The location of the intervertebral disc herniation in French Bulldogs with EH was more likely to be thoracolumbar (OR: 10.8; 95% CI: 2.1-55.7; P = .03). CONCLUSIONS AND CLINICAL IMPORTANCE: French Bulldogs have a high prevalence of intervertebral disc-associated EH. Dogs with EH have a shorter clinical course and are more likely to be nonambulatory on initial presentation.


Assuntos
Doenças do Cão , Deslocamento do Disco Intervertebral , Disco Intervertebral , Compressão da Medula Espinal , Animais , Estudos Transversais , Doenças do Cão/diagnóstico por imagem , Doenças do Cão/epidemiologia , Doenças do Cão/cirurgia , Cães , Hemorragia/veterinária , Deslocamento do Disco Intervertebral/diagnóstico por imagem , Deslocamento do Disco Intervertebral/cirurgia , Deslocamento do Disco Intervertebral/veterinária , Prevalência , Estudos Retrospectivos , Compressão da Medula Espinal/diagnóstico por imagem , Compressão da Medula Espinal/cirurgia , Compressão da Medula Espinal/veterinária
14.
Front Vet Sci ; 9: 888483, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35664857

RESUMO

Sorafenib is a multi-kinase small molecule inhibitor that targets serine/threonine and tyrosine kinases including the RAF kinase family, VEGFR-2, and PDGFR. The aim of this study was to evaluate the systemic pharmacokinetics of a previously defined tolerable oral dose of sorafenib in tumor-bearing dogs. Six client-owned dogs with a cytologic or histologic diagnosis of cancer were enrolled in this open-label, tolerability study. Dogs were administered sorafenib at an intended dose of 3 mg/kg and serum samples were obtained for analysis of sorafenib serum concentrations at 0, 1, 2, 6, 12, 24, 48, 72, 96, and 168 h post-drug administration. Median time to peak serum sorafenib concentration occurred at 4 h (range 2-12 h) resulting in an average serum concentration of 54.9 ± 33.5 ng/mL (118.2 ± 72.1 nM). Mean sorafenib levels declined by over 70% relative to peak serum concentrations by 24 h in all dogs, suggesting the value of at least twice daily administration. Doses of 3 mg/kg were well-tolerated and no patients in the study experienced adverse events that were attributable to sorafenib. Future trials in dogs with cancer are recommended at this dosing schedule to assess the effect of sorafenib administration on anti-tumor efficacy signals and relevant pharmacodynamic target modulation in vivo.

15.
J Vet Intern Med ; 36(4): 1382-1389, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35621070

RESUMO

BACKGROUND: Necrotizing meningoencephalitis (NME) in the pug dogs is a fatal neuroinflammatory disease associated with rapid progression and poor response to conventional immunosuppressive therapy. Diagnosis is typically made after severe neurological abnormalities have manifested. HYPOTHESIS/OBJECTIVE: Pug dogs at genetic risk for NME might manifest neurological abnormalities before developing pathognomonic clinical signs of NME. ANIMALS: Thirty-six pug dogs less than 4 years of age asymptomatic for NME. METHODS: Prospective observational cohort study with germline genome-wide genotyping. Neurological examinations were performed 4 weeks apart to document reproducible findings of central nervous system disease. Magnetic resonance imaging, cerebrospinal fluid analysis, and testing for infectious diseases were performed in all pugs with reproducible abnormalities detected on neurological examination. RESULTS: The overall risk allele frequency in this cohort was 40%; 5 (14%) dogs were high risk, 19 (53%) dogs were medium risk, and 12 (33%) dogs were low genetic risk for NME. Reproducible abnormalities detected on neurological examination were identified in 8/24 (33%) genetically at-risk dogs and 0/12 (0%) low risk dogs. Clinical abnormalities included multifocal spinal pain in 8/8, reduced menace response in 5/8, and lateralizing postural reaction deficits in 5/8 pugs. There was a strong association between genotype risk and the presence of this clinical phenotype (P = .03). CONCLUSIONS AND CLINICAL IMPORTANCE: Our findings suggest the presence of a novel early clinical phenotype of NME in apparently asymptomatic genetically at-risk pugs which might be used to plan early diagnostic and therapeutic clinical trials.


Assuntos
Doenças do Cão , Meningoencefalite , Animais , Cães , Doenças do Cão/líquido cefalorraquidiano , Doenças do Cão/diagnóstico , Doenças do Cão/genética , Frequência do Gene , Meningoencefalite/líquido cefalorraquidiano , Meningoencefalite/genética , Meningoencefalite/veterinária , Fenótipo , Estudos Prospectivos
16.
Sci Rep ; 12(1): 6337, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35428782

RESUMO

Comparative studies of naturally occurring canine cancers have provided new insight into many areas of cancer research. Development and validation of circulating tumor DNA (ctDNA) analysis in pet dogs can help address diagnostic needs in veterinary as well as human oncology. Dogs have high incidence of naturally occurring spontaneous cancers, demonstrate molecular heterogeneity and clonal evolution during therapy, allow serial sampling of blood from the same individuals during the course of disease progression, and have relatively compressed intervals for disease progression amenable to longitudinal studies. Here, we present a feasibility study of ctDNA analysis performed in 48 dogs including healthy dogs and dogs with either benign splenic lesions or malignant splenic tumors (hemangiosarcoma) using shallow whole genome sequencing (sWGS) of cell-free DNA. To enable detection and quantification of ctDNA using sWGS, we adapted two informatic approaches and compared their performance for the canine genome. At the time of initial clinical presentation, mean ctDNA fraction in dogs with malignant splenic tumors was 11.2%, significantly higher than dogs with benign lesions (3.2%; p = 0.001). ctDNA fraction was 14.3% and 9.0% in dogs with metastatic and localized disease, respectively (p = 0.227). In dogs treated with surgical resection of malignant tumors, mean ctDNA fraction decreased from 11.0% prior to resection to 7.9% post-resection (p = 0.047 for comparison of paired samples). Our results demonstrate that ctDNA analysis is feasible in dogs with hemangiosarcoma using a cost-effective approach such as sWGS. Additional studies are needed to validate these findings, and determine the role of ctDNA to assess burden of disease and treatment response in dogs with cancer.


Assuntos
DNA Tumoral Circulante , Hemangiossarcoma , Neoplasias Esplênicas , Animais , Biomarcadores Tumorais/genética , DNA Tumoral Circulante/genética , Progressão da Doença , Cães , Estudos de Viabilidade , Hemangiossarcoma/genética , Hemangiossarcoma/veterinária , Mutação , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/veterinária
17.
PLoS One ; 17(7): e0264986, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35867969

RESUMO

Cancer genomic heterogeneity presents significant challenges for understanding oncogenic processes and for cancer's clinical management. Variation in driver mutation frequency between patients with the same tumor type as well as within an individual patients' cancer can shape the use of mutations as diagnostic, prognostic, and predictive biomarkers. We have characterized genomic heterogeneity between and within canine splenic hemangiosarcoma (HSA), a common naturally occurring cancer in pet dogs that is similar to human angiosarcoma (AS). HSA is a clinically, physiologically, and genomically complex canine cancer that may serve as a valuable model for understanding the origin and clinical impact of cancer heterogeneity. We conducted a prospective collection of 52 splenic masses from 43 dogs (27 HSA, 15 benign masses, and 1 stromal sarcoma) presenting for emergency care with hemoperitoneum secondary to a ruptured splenic mass. Multi-platform genomic analysis included matched tumor/normal targeted sequencing panel and exome sequencing. We found candidate somatic cancer driver mutations in 14/27 (52%) HSAs. Among recurrent candidate driver mutations, TP53 was most commonly mutated (30%) followed by PIK3CA (15%), AKT1 (11%), and CDKN2AIP (11%). We also identified significant intratumoral genomic heterogeneity, consistent with a branched evolution model, through multi-region exome sequencing of three distinct tumor regions from selected primary splenic tumors. These data provide new perspectives on the genomic landscape of this veterinary cancer and suggest a cross-species value for using HSA in pet dogs as a naturally occurring model of intratumoral heterogeneity.


Assuntos
Doenças do Cão , Hemangiossarcoma , Neoplasias Esplênicas , Animais , Doenças do Cão/genética , Cães , Genômica , Hemangiossarcoma/genética , Hemangiossarcoma/veterinária , Humanos , Mutação , Estudos Prospectivos , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/veterinária , Sequenciamento do Exoma
18.
Nat Med ; 10(2): 175-81, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14704789

RESUMO

Patients presenting with metastatic rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children, have a very poor clinical prognosis. This is due, in large part, to our rudimentary knowledge of the molecular events that dictate metastatic potential. We used cDNA microarray analysis of RMS cell lines, derived from Ink4a/Arf-deficient mice transgenic for hepatocyte growth factor/scatter factor (HGF/SF), to identify a set of genes whose expression was significantly different between highly and poorly metastatic cells. Subsequent in vivo functional studies revealed that the actin filament-plasma membrane linker ezrin (encoded by Vil2) and the homeodomain-containing transcription factor Six-1 (sine oculis-related homeobox-1 homolog) had essential roles in determining the metastatic fate of RMS cells. VIL2 and SIX1 expression was enhanced in human RMS tissue, significantly correlating with clinical stage. The identification of ezrin and Six-1 as critical regulators of metastasis in RMS provides new mechanistic and therapeutic insights into this pediatric cancer.


Assuntos
Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Metástase Neoplásica , Fosfoproteínas/metabolismo , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/secundário , Animais , Linhagem Celular Tumoral , Criança , Proteínas do Citoesqueleto , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fosfoproteínas/genética , Filogenia , Rabdomiossarcoma/patologia
19.
Nat Med ; 10(2): 182-6, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14704791

RESUMO

Metastatic cancers, once established, are the primary cause of mortality associated with cancer. Previously, we used a genomic approach to identify metastasis-associated genes in cancer. From this genomic data, we selected ezrin for further study based on its role in physically and functionally connecting the actin cytoskeleton to the cell membrane. In a mouse model of osteosarcoma, a highly metastatic pediatric cancer, we found ezrin to be necessary for metastasis. By imaging metastatic cells in the lungs of mice, we showed that ezrin expression provided an early survival advantage for cancer cells that reached the lung. AKT and MAPK phosphorylation and activity were reduced when ezrin protein was suppressed. Ezrin-mediated early metastatic survival was partially dependent on activation of MAPK, but not AKT. To define the relevance of ezrin in the biology of metastasis, beyond the founding mouse model, we examined ezrin expression in dogs that naturally developed osteosarcoma. High ezrin expression in dog tumors was associated with early development of metastases. Consistent with this data, we found a significant association between high ezrin expression and poor outcome in pediatric osteosarcoma patients.


Assuntos
Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Metástase Neoplásica , Osteossarcoma/metabolismo , Osteossarcoma/secundário , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Linhagem Celular Tumoral , Criança , Proteínas do Citoesqueleto , Cães , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/fisiologia , Taxa de Sobrevida
20.
J Pediatr Hematol Oncol ; 33(7): 524-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21941146

RESUMO

The ability to define osteosarcoma (OS) patients at greatest risk for metastatic progression and nonresponsiveness to conventional therapy is currently not possible. Such biomarkers are needed to predict overall prognosis, probability of metastases at diagnosis, and response to chemotherapy. The tissue microarray (TMA) serves as a powerful tool for detecting and validating protein biomarkers across a variety of patients. We constructed a novel outcome-linked TMA to add to and address shortcomings of currently available OS tissue resources. To test the use of our TMA, we surveyed the expression of eukaryotic initiation factor 4E (eIF4E) in OS patients using immunohistochemistry. Aberrant regulation of translation initiation is a feature of many cancers. eIF4E is central to initiation of protein synthesis. Its expression and activity have been implicated in tumor formation and potentially malignant and/or metastatic progression in some carcinomas. We found that eIF4E was uniformly expressed in OS patient samples. No association was found between eIF4E and outcome in OS patients. This novel OS TMA provided a facile mechanism to assess the role of a relevant protein biomarker in OS.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias Ósseas/metabolismo , Fator de Iniciação 4E em Eucariotos/biossíntese , Osteossarcoma/metabolismo , Análise Serial de Tecidos , Adolescente , Adulto , Neoplasias Ósseas/diagnóstico , Neoplasias Ósseas/terapia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Osteossarcoma/diagnóstico , Osteossarcoma/terapia , Taxa de Sobrevida , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA