Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cerebellum ; 22(6): 1166-1181, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36242761

RESUMEN

Essential tremor (ET) is a common, progressive neurological disease characterized by an 8-12-Hz kinetic tremor. Despite its high prevalence, the patho-mechanisms of tremor in ET are not fully known. Through comprehensive studies in postmortem brains, we identified major morphological changes in the ET cerebellum that reflect cellular damage in Purkinje cells (PCs), suggesting that PC damage is central to ET pathogenesis. We previously performed a transcriptome analysis in ET cerebellar cortex, identifying candidate genes and several dysregulated pathways. To directly target PCs, we purified RNA from PCs isolated by laser capture microdissection and performed the first ever PC-specific RNA-sequencing analysis in ET versus controls. Frozen postmortem cerebellar cortex from 24 ETs and 16 controls underwent laser capture microdissection, obtaining ≥2000 PCs per sample. RNA transcriptome was analyzed via differential gene expression, principal component analysis (PCA), and gene set enrichment analyses (GSEA). We identified 36 differentially expressed genes, encompassing multiple cellular processes. Some ET (13/24) had greater dysregulation of these genes and segregated from most controls and remaining ETs in PCA. Characterization of genes/pathways enriched in this PCA and GSEA identified multiple pathway dysregulations in ET, including RNA processing/splicing, synapse organization/ion transport, and oxidative stress/inflammation. Furthermore, a different set of pathways characterized marked heterogeneity among ET patients. Our data indicate a range of possible mechanisms for the pathogenesis of ET. Significant heterogeneity among ET combined with dysregulation of multiple cellular processes supports the notion that ET is a family of disorders rather than one disease entity.


Asunto(s)
Temblor Esencial , Células de Purkinje , Humanos , Células de Purkinje/metabolismo , Temblor Esencial/patología , Temblor/patología , Cerebelo/patología , Perfilación de la Expresión Génica , ARN/metabolismo , Rayos Láser
2.
PLoS One ; 17(7): e0264986, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35867969

RESUMEN

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.


Asunto(s)
Enfermedades de los Perros , Hemangiosarcoma , Neoplasias del Bazo , Animales , Enfermedades de los Perros/genética , Perros , Genómica , Hemangiosarcoma/genética , Hemangiosarcoma/veterinaria , Humanos , Mutación , Estudios Prospectivos , Neoplasias del Bazo/genética , Neoplasias del Bazo/veterinaria , Secuenciación del Exoma
3.
Artículo en Inglés | MEDLINE | ID: mdl-34848502

RESUMEN

BACKGROUND AND OBJECTIVES: Patients with Alzheimer dementia display evidence of amyloid-related neurodegeneration. Our focus was to determine whether such patients also display evidence of a disease-targeting adaptive immune response mediated by CD4+ T cells. To test this hypothesis, we evaluated the CSF immune profiles of patients with Alzheimer clinical syndrome (ACS), who display clinically defined dementia. METHODS: Innate and adaptive immune profiles of patients with ACS were measured using multicolor flow cytometry. CSF-derived CD4+ and CD8+ T-cell receptor repertoire genetics were measured using next-generation sequencing. Brain-specific autoantibody signatures of CSF-derived antibody pools were measured using array technology or ELISA. CSF from similar-age healthy controls (HCs) was used as a comparator cohort. RESULTS: Innate cells were expanded in the CSF of patients with ACS in comparison to HCs, and innate cell expansion increased with age in the patients with ACS, but not HCs. Despite innate cell expansion in the CSF, the frequency of total CD4+ T cells reduced with age in the patients with ACS. T-cell receptor repertoire genetics indicated that T-cell clonal expansion is enhanced, and diversity is reduced in the patients with ACS compared with similar-age HCs. DISCUSSION: Examination of CSF indicates that CD4+ T cell-mediated adaptive immune responses are altered in patients with ACS. Understanding the underlying mechanisms affecting adaptive immunity will help move us toward the goal of slowing cognitive decline.


Asunto(s)
Inmunidad Adaptativa/inmunología , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/inmunología , Autoanticuerpos/líquido cefalorraquídeo , Linfocitos T CD4-Positivos/metabolismo , Inmunidad Innata/inmunología , Anciano , Linfocitos T CD8-positivos/metabolismo , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndrome
4.
Elife ; 102021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34473622

RESUMEN

Concussion is associated with a myriad of deleterious immediate and long-term consequences. Yet the molecular mechanisms and genetic targets promoting the selective vulnerability of different neural subtypes to dysfunction and degeneration remain unclear. Translating experimental models of blunt force trauma in C. elegans to concussion in mice, we identify a conserved neuroprotective mechanism in which reduction of mitochondrial electron flux through complex IV suppresses trauma-induced degeneration of the highly vulnerable dopaminergic neurons. Reducing cytochrome C oxidase function elevates mitochondrial-derived reactive oxygen species, which signal through the cytosolic hypoxia inducing transcription factor, Hif1a, to promote hyperphosphorylation and inactivation of the pyruvate dehydrogenase, PDHE1α. This critical enzyme initiates the Warburg shunt, which drives energetic reallocation from mitochondrial respiration to astrocyte-mediated glycolysis in a neuroprotective manner. These studies demonstrate a conserved process in which glycolytic preconditioning suppresses Parkinson-like hypersensitivity of dopaminergic neurons to trauma-induced degeneration via redox signaling and the Warburg effect.


Concussion is a type of traumatic brain injury that results from a sudden blow or jolt to the head. Symptoms can include a passing headache, dizziness, confusion or sensitivity to light, but experiencing multiple concussions can have drastic repercussions in later life. Studies of professional athletes have shown that those who experience one or more concussions are prone to developing Alzheimer's and Parkinson's disease, two well-known neurodegenerative diseases. Both conditions involve the progressive loss or breakdown of nerve cells, called neurons. But exactly how this so-called neurodegeneration of brain cells stems from the original, physical injury remains unclear. Head trauma may cause damage to the structural support of a cell or disrupt the flow of electrical impulses through neurons. Energy use and production in damaged cells could shift into overdrive to repair the damage. The chemical properties of different types of brain cells could also make some more vulnerable to trauma than others. Besides neurons, star-shaped support cells in the brain called astrocytes, which may have some protective ability, could also be affected. To investigate which cells may be more susceptible to traumatic injuries, Solano Fonseca et al. modelled the impacts of concussion-like head trauma in roundworms (C. elegans) and mice. In both animals, one type of neuron was extremely vulnerable to cell death after trauma. Neurons that release dopamine, a chemical involved in cell-to-cell communication and the brain's reward system, showed signs of cell damage and deteriorated after injury. Dopaminergic cells, as these cells are called, are involved in motor coordination, and the loss of dopaminergic cells has been linked to both Alzheimer's and Parkinson's disease. Astrocytes, however, had a role in reducing the death of dopaminergic neurons after trauma. In experiments, astrocytes appeared to restore the balance of energy production to meet the increased energy demands of impacted neurons. Single-cell analyses showed that genes involved in metabolism were switched on in astrocytes to produce energy via an alternative pathway. This energetic shift facilitated via astrocytes may help mitigate against some damage to dopamine-producing neurons after trauma, reducing cell death. This work furthers our understanding of cellular changes in the concussed brain. More research will be required to better characterise how this immediate trauma to cells, and the subsequent loss of dopaminergic neurons, impacts brain health long-term. Efforts to design effective therapies to slow or reverse these changes could then follow.


Asunto(s)
Astrocitos , Lesiones Traumáticas del Encéfalo , Glucólisis/fisiología , Degeneración Nerviosa , Neuroprotección/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Caenorhabditis elegans , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Células HEK293 , Humanos , Ratones , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología
5.
Neuron ; 109(13): 2091-2105.e6, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34051145

RESUMEN

The hippocampus supports many facets of cognition, including learning, memory, and emotional processing. Anatomically, the hippocampus runs along a longitudinal axis, posterior to anterior in primates. The structure, function, and connectivity of the hippocampus vary along this axis. In human hippocampus, longitudinal functional heterogeneity remains an active area of investigation, and structural heterogeneity has not been described. To understand the cellular and molecular diversity along the hippocampal long axis in human brain and define molecular signatures corresponding to functional domains, we performed single-nuclei RNA sequencing on surgically resected human anterior and posterior hippocampus from epilepsy patients, identifying differentially expressed genes at cellular resolution. We further identify axis- and cell-type-specific gene expression signatures that differentially intersect with human genetic signals, identifying cell-type-specific genes in the posterior hippocampus for cognitive function and the anterior hippocampus for mood and affect. These data are accessible as a public resource through an interactive website.


Asunto(s)
Expresión Génica , Hipocampo/metabolismo , Neuronas/metabolismo , Femenino , Perfilación de la Expresión Génica , Hipocampo/anatomía & histología , Humanos , Masculino , Microglía/metabolismo , Análisis de Secuencia de ARN
6.
PLoS One ; 16(4): e0248097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33826614

RESUMEN

Although combination BRAF and MEK inhibitors are highly effective for the 40-50% of cutaneous metastatic melanomas harboring BRAFV600 mutations, targeted agents have been ineffective for BRAFV600wild-type (wt) metastatic melanomas. The SU2C Genomics-Enabled Medicine for Melanoma Trial utilized a Simon two-stage optimal design to assess whether comprehensive genomic profiling improves selection of molecular-based therapies for BRAFV600wt metastatic melanoma patients who had progressed on standard-of-care therapy, which may include immunotherapy. Of the response-evaluable patients, binimetinib was selected for 20 patients randomized to the genomics-enabled arm, and nine were treated on the alternate treatment arm. Response rates for 27 patients treated with targeted recommendations included one (4%) partial response, 18 (67%) with stable disease, and eight (30%) with progressive disease. Post-trial genomic and protein pathway activation mapping identified additional drug classes that may be considered for future studies. Our results highlight the complexity and heterogeneity of metastatic melanomas, as well as how the lack of response in this trial may be associated with limitations including monotherapy drug selection and the dearth of available single and combination molecularly-driven therapies to treat BRAFV600wt metastatic melanomas.


Asunto(s)
Bencimidazoles/administración & dosificación , Genómica , Melanoma , Proteínas Proto-Oncogénicas B-raf , Neoplasias Cutáneas , Adulto , Anciano , Femenino , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Persona de Mediana Edad , Metástasis de la Neoplasia , Proyectos Piloto , Estudios Prospectivos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Melanoma Cutáneo Maligno
7.
Clin Cancer Res ; 25(19): 5866-5877, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31431454

RESUMEN

PURPOSE: Naturally occurring primary canine lung cancers share clinicopathologic features with human lung cancers in never-smokers, but the genetic underpinnings of canine lung cancer are unknown. We have charted the genomic landscape of canine lung cancer and performed functional characterization of novel, recurrent HER2 (ERBB2) mutations occurring in canine pulmonary adenocarcinoma (cPAC). EXPERIMENTAL DESIGN: We performed multiplatform genomic sequencing of 88 primary canine lung tumors or cell lines. Additionally, in cPAC cell lines, we performed functional characterization of HER2 signaling and evaluated mutation-dependent HER2 inhibitor drug dose-response. RESULTS: We discovered somatic, coding HER2 point mutations in 38% of cPACs (28/74), but none in adenosquamous (cPASC, 0/11) or squamous cell (cPSCC, 0/3) carcinomas. The majority (93%) of HER2 mutations were hotspot V659E transmembrane domain (TMD) mutations comparable to activating mutations at this same site in human cancer. Other HER2 mutations were located in the extracellular domain and TMD. HER2 V659E was detected in the plasma of 33% (2/6) of dogs with localized HER2 V659E tumors. HER2 V659E cPAC cell lines displayed constitutive phosphorylation of AKT and significantly higher sensitivity to the HER2 inhibitors lapatinib and neratinib relative to HER2-wild-type cell lines (IC50 < 200 nmol/L in HER2 V659E vs. IC50 > 2,500 nmol/L in HER2 WT). CONCLUSIONS: This study creates a foundation for molecular understanding of and drug development for canine lung cancer. These data also establish molecular contexts for comparative studies in dogs and humans of low mutation burden, never-smoker lung cancer, and mutant HER2 function and inhibition.


Asunto(s)
Adenocarcinoma del Pulmón/veterinaria , Enfermedades de los Perros/genética , Neoplasias Pulmonares/veterinaria , Mutación , Receptor ErbB-2/genética , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Supervivencia Celular/efectos de los fármacos , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/patología , Perros , Femenino , Lapatinib/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Transducción de Señal , Células Tumorales Cultivadas
8.
Commun Biol ; 2: 266, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31341965

RESUMEN

Osteosarcoma (OS) is a rare, metastatic, human adolescent cancer that also occurs in pet dogs. To define the genomic underpinnings of canine OS, we performed multi-platform analysis of OS tumors from 59 dogs, including whole genome sequencing (n = 24) and whole exome sequencing (WES; n = 13) of primary tumors and matched normal tissue, WES (n = 10) of matched primary/metastatic/normal samples and RNA sequencing (n = 54) of primary tumors. We found that canine OS recapitulates features of human OS including low point mutation burden (median 1.98 per Mb) with a trend towards higher burden in metastases, high structural complexity, frequent TP53 (71%), PI3K pathway (37%), and MAPK pathway mutations (17%), and low expression of immune-associated genes. We also identified novel features of canine OS including putatively inactivating somatic SETD2 (42%) and DMD (50%) aberrations. These findings set the stage for understanding OS development in dogs and humans, and establish genomic contexts for future comparative analyses.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/veterinaria , Distrofina/genética , N-Metiltransferasa de Histona-Lisina/genética , Mutación , Osteosarcoma/genética , Osteosarcoma/veterinaria , Animales , Perros , Secuenciación Completa del Genoma
9.
PLoS Genet ; 14(9): e1007589, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30188888

RESUMEN

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.


Asunto(s)
Melanoma/genética , Melanoma/veterinaria , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/veterinaria , Animales , Ciclo Celular/genética , Proliferación Celular/genética , Hibridación Genómica Comparativa , Análisis Mutacional de ADN , Enfermedades de los Perros/genética , Perros , Femenino , Masculino , Melanoma/sangre , Melanoma/patología , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Transducción de Señal/genética , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/patología , Análisis de Matrices Tisulares
10.
Genome Res ; 27(4): 524-532, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28373299

RESUMEN

Genomic analyses of cutaneous melanoma (CM) have yielded biological and therapeutic insights, but understanding of non-ultraviolet (UV)-derived CMs remains limited. Deeper analysis of acral lentiginous melanoma (ALM), a rare sun-shielded melanoma subtype associated with worse survival than CM, is needed to delineate non-UV oncogenic mechanisms. We thus performed comprehensive genomic and transcriptomic analysis of 34 ALM patients. Unlike CM, somatic alterations were dominated by structural variation and absence of UV-derived mutation signatures. Only 38% of patients demonstrated driver BRAF/NRAS/NF1 mutations. In contrast with CM, we observed PAK1 copy gains in 15% of patients, and somatic TERT translocations, copy gains, and missense and promoter mutations, or germline events, in 41% of patients. We further show that in vitro TERT inhibition has cytotoxic effects on primary ALM cells. These findings provide insight into the role of TERT in ALM tumorigenesis and reveal preliminary evidence that TERT inhibition represents a potential therapeutic strategy in ALM.


Asunto(s)
Aberraciones Cromosómicas , Melanoma/genética , Mutación , Neoplasias Cutáneas/genética , Telomerasa/genética , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Femenino , GTP Fosfohidrolasas/genética , Genes de Neurofibromatosis 1 , Humanos , Masculino , Melanoma/patología , Proteínas de la Membrana/genética , Persona de Mediana Edad , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/patología , Telomerasa/metabolismo , Transcriptoma , Quinasas p21 Activadas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...