Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 32(8): 1961-1973, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34045313

RESUMEN

BACKGROUND: Mutations in COL4A5 are responsible for 80% of cases of X-linked Alport Syndrome (XLAS). Although genes that cause AS are well characterized, people with AS who have similar genetic mutations present with a wide variation in the extent of kidney impairment and age of onset, suggesting the activities of modifier genes. METHODS: We created a cohort of genetically diverse XLAS male and female mice using the Diversity Outbred mouse resource and measured albuminuria, GFR, and gene expression. Using a quantitative trait locus approach, we mapped modifier genes that can best explain the underlying phenotypic variation measured in our diverse population. RESULTS: Genetic analysis identified several loci associated with the variation in albuminuria and GFR, including a locus on the X chromosome associated with X inactivation and a locus on chromosome 2 containing Fmn1. Subsequent analysis of genetically reduced Fmn1 expression in Col4a5 knockout mice showed a decrease in albuminuria, podocyte effacement, and podocyte protrusions in the glomerular basement membrane, which support the candidacy of Fmn1 as a modifier gene for AS. CONCLUSION: With this novel approach, we emulated the variability in the severity of kidney phenotypes found in human patients with Alport Syndrome through albuminuria and GFR measurements. This approach can identify modifier genes in kidney disease that can be used as novel therapeutic targets.


Asunto(s)
Albuminuria/orina , Colágeno Tipo IV/genética , Creatinina/orina , Forminas/genética , Nefritis Hereditaria/genética , Albuminuria/etiología , Animales , Mapeo Cromosómico , Modelos Animales de Enfermedad , Femenino , Forminas/ultraestructura , Expresión Génica , Tasa de Filtración Glomerular , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mutación , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/fisiopatología , Fenotipo , Podocitos/patología , Prueba de Estudio Conceptual , Sitios de Carácter Cuantitativo , RNA-Seq , Factores Sexuales , Secuenciación Completa del Genoma
2.
J Am Soc Nephrol ; 32(1): 52-68, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33154175

RESUMEN

BACKGROUND: Nephropathologic analyses provide important outcomes-related data in experiments with the animal models that are essential for understanding kidney disease pathophysiology. Precision medicine increases the demand for quantitative, unbiased, reproducible, and efficient histopathologic analyses, which will require novel high-throughput tools. A deep learning technique, the convolutional neural network, is increasingly applied in pathology because of its high performance in tasks like histology segmentation. METHODS: We investigated use of a convolutional neural network architecture for accurate segmentation of periodic acid-Schiff-stained kidney tissue from healthy mice and five murine disease models and from other species used in preclinical research. We trained the convolutional neural network to segment six major renal structures: glomerular tuft, glomerulus including Bowman's capsule, tubules, arteries, arterial lumina, and veins. To achieve high accuracy, we performed a large number of expert-based annotations, 72,722 in total. RESULTS: Multiclass segmentation performance was very high in all disease models. The convolutional neural network allowed high-throughput and large-scale, quantitative and comparative analyses of various models. In disease models, computational feature extraction revealed interstitial expansion, tubular dilation and atrophy, and glomerular size variability. Validation showed a high correlation of findings with current standard morphometric analysis. The convolutional neural network also showed high performance in other species used in research-including rats, pigs, bears, and marmosets-as well as in humans, providing a translational bridge between preclinical and clinical studies. CONCLUSIONS: We developed a deep learning algorithm for accurate multiclass segmentation of digital whole-slide images of periodic acid-Schiff-stained kidneys from various species and renal disease models. This enables reproducible quantitative histopathologic analyses in preclinical models that also might be applicable to clinical studies.


Asunto(s)
Aprendizaje Profundo , Diagnóstico por Computador , Riñón/fisiopatología , Reconocimiento de Normas Patrones Automatizadas , Algoritmos , Animales , Modelos Animales de Enfermedad , Procesamiento de Imagen Asistido por Computador/métodos , Enfermedades Renales/patología , Glomérulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Redes Neurales de la Computación , Ácido Peryódico/química , Reproducibilidad de los Resultados , Bases de Schiff , Investigación Biomédica Traslacional
3.
Am J Pathol ; 189(9): 1786-1796, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31220455

RESUMEN

With the advent and increased accessibility of deep neural networks (DNNs), complex properties of histologic images can be rigorously and reproducibly quantified. We used DNN-based transfer learning to analyze histologic images of periodic acid-Schiff-stained renal sections from a cohort of mice with different genotypes. We demonstrate that DNN-based machine learning has strong generalization performance on multiple histologic image processing tasks. The neural network extracted quantitative image features and used them as classifiers to look for differences between mice of different genotypes. Excellent performance was observed at segmenting glomeruli from non-glomerular structure and subsequently predicting the genotype of the animal on the basis of glomerular quantitative image features. The DNN-based genotype classifications highly correlate with mesangial matrix expansion scored by a pathologist (R.E.C.), which differed in these animals. In addition, by analyzing non-glomeruli images, the neural network identified novel histologic features that differed by genotype, including the presence of vacuoles, nuclear count, and proximal tubule brush border integrity, which was validated with immunohistologic staining. These features were not identified in systematic pathologic examination. Our study demonstrates the power of DNNs to extract biologically relevant phenotypes and serve as a platform for discovering novel phenotypes. These results highlight the synergistic possibilities for pathologists and DNNs to radically scale up our ability to generate novel mechanistic hypotheses in disease.


Asunto(s)
Aldehído Oxidorreductasas/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Riñón/fisiopatología , Redes Neurales de la Computación , Vías Nerviosas , Animales , Masculino , Ratones , Ratones Noqueados , Fenotipo
4.
Hum Mol Genet ; 26(19): 3722-3735, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28934385

RESUMEN

Mutations of the human ATP6V1B1 gene cause distal renal tubular acidosis (dRTA; OMIM #267300) often associated with sensorineural hearing impairment; however, mice with a knockout mutation of Atp6v1b1 were reported to exhibit a compensated acidosis and normal hearing. We discovered a new spontaneous mutation (vortex, symbol vtx) of Atp6v1b1 in an MRL/MpJ (MRL) colony of mice. In contrast to the reported phenotype of the knockout mouse, which was developed on a primarily C57BL/6 (B6) strain background, MRL-Atp6v1b1vtx/vtx mutant mice exhibit profound hearing impairment, which is associated with enlarged endolymphatic compartments of the inner ear. Mutant mice have alkaline urine but do not exhibit overt metabolic acidosis, a renal phenotype similar to that of the Atpbv1b1 knockout mouse. The abnormal inner ear phenotype of MRL- Atp6v1b1vtx/vtx mice was lost when the mutation was transferred onto the C57BL/6J (B6) background, indicating the influence of strain-specific genetic modifiers. To genetically map modifier loci in Atp6v1b1vtx/vtx mice, we analysed ABR thresholds of progeny from a backcross segregating MRL and B6 alleles. We found statistically significant linkage with a locus on Chr 13 that accounts for about 20% of the hearing threshold variation in the backcross mice. The important effect that genetic background has on the inner ear phenotype of Atp6v1b1 mutant mice provides insight into the hearing loss variability associated with dRTA caused by ATP6V1B1 mutations. Because MRL-Atp6v1b1vxt/vtx mice do not recapitulate the metabolic acidosis of dRTA patients, they provide a new genetic model for nonsyndromic deafness with enlarged vestibular aqueduct (EVA; OMIM #600791).


Asunto(s)
Sordera/genética , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Acidosis/genética , Acidosis/metabolismo , Acidosis Tubular Renal/genética , Acidosis Tubular Renal/metabolismo , Animales , Sordera/metabolismo , Modelos Animales de Enfermedad , Oído Interno/patología , Femenino , Ligamiento Genético , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Acueducto Vestibular/metabolismo , Acueducto Vestibular/fisiología
5.
Physiol Genomics ; 50(8): 543-552, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29652635

RESUMEN

Mesangial matrix expansion is an important process in the initiation of chronic kidney disease, yet the genetic factors driving its development are unknown. Our previous studies have implicated Far2 as a candidate gene associated with differences in mesangial matrix expansion between mouse inbred strains. Consistent with the hypothesis that increased expression of Far2 leads to mesangial matrix expansion through increased production of platelet-activating factor precursors, we show that FAR2 is capable of mediating de novo platelet-activating factor synthesis in vitro and driven by the transcription factor NKX3.2. We demonstrate that knockdown of Far2 in mice delays the progression of mesangial matrix expansion with at least six months (equivalent to ~15 yr in human). Furthermore, we show that increased FAR2 expression in human patients is associated with diabetic nephropathy, lupus nephritis, and IgA nephropathy. Taken together, these results highlight FAR2's role in the development of mesangial matrix expansion and chronic kidney disease.


Asunto(s)
Aldehído Oxidorreductasas/genética , Nefropatías Diabéticas/genética , Mesangio Glomerular/metabolismo , Adulto , Anciano , Aldehído Oxidorreductasas/metabolismo , Animales , Nefropatías Diabéticas/metabolismo , Femenino , Mesangio Glomerular/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto Joven
6.
Am J Physiol Renal Physiol ; 315(6): F1644-F1651, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30256126

RESUMEN

Current methods of scoring histological kidney samples, specifically glomeruli, do not allow for collection of quantitative data in a high-throughput and consistent manner. Neither untrained individuals nor computers are presently capable of identifying glomerular features, so expert pathologists must do the identification and score using a categorical matrix, complicating statistical analysis. Critical information regarding overall health and physiology is encoded in these samples. Rapid comprehensive histological scoring could be used, in combination with other physiological measures, to significantly advance renal research. Therefore, we used machine learning to develop a high-throughput method to automatically identify and collect quantitative data from glomeruli. Our method requires minimal human interaction between steps and provides quantifiable data independent of user bias. The method uses free existing software and is usable without extensive image analysis training. Validation of the classifier and feature scores in mice is highlighted in this work and shows the power of applying this method in murine research. Preliminary results indicate that the method can be applied to data sets from different species after training on relevant data, allowing for fast glomerular identification and quantitative measurements of glomerular features. Validation of the classifier and feature scores are highlighted in this work and show the power of applying this method. The resulting data are free from user bias. Continuous data, such that statistical analysis can be performed, allows for more precise and comprehensive interrogation of samples. These data can then be combined with other physiological data to broaden our overall understanding of renal function.


Asunto(s)
Diagnóstico por Computador/métodos , Interpretación de Imagen Asistida por Computador/métodos , Enfermedades Renales/patología , Glomérulos Renales/patología , Aprendizaje Automático , Microscopía/métodos , Reconocimiento de Normas Patrones Automatizadas/métodos , Animales , Automatización de Laboratorios , Biopsia , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Valor Predictivo de las Pruebas , Ratas , Reproducibilidad de los Resultados , Programas Informáticos , Especificidad de la Especie , Flujo de Trabajo
7.
J Am Soc Nephrol ; 27(11): 3271-3277, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27020856

RESUMEN

Changes in metabolite levels of the kynurenine pathway have been observed in patients with CKD, suggesting involvement of this pathway in disease pathogenesis. Our recent genetic analysis in the mouse identified the kynurenine 3-mono-oxygenase (KMO) gene (Kmo) as a candidate gene associated with albuminuria. This study investigated this association in more detail. We compared KMO abundance in the glomeruli of mice and humans under normal and diabetic conditions, observing a decrease in glomerular KMO expression with diabetes. Knockdown of kmo expression in zebrafish and genetic deletion of Kmo in mice each led to a proteinuria phenotype. We observed pronounced podocyte foot process effacement on long stretches of the filtration barrier in the zebrafish knockdown model and mild podocyte foot process effacement in the mouse model, whereas all other structures within the kidney remained unremarkable. These data establish the candidacy of KMO as a causal factor for changes in the kidney leading to proteinuria and indicate a functional role for KMO and metabolites of the tryptophan pathway in podocytes.


Asunto(s)
Eliminación de Gen , Quinurenina 3-Monooxigenasa/genética , Proteinuria/enzimología , Proteinuria/genética , Animales , Femenino , Humanos , Quinurenina 3-Monooxigenasa/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Pez Cebra
8.
Invest New Drugs ; 33(1): 128-37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25248753

RESUMEN

BACKGROUND: Ganetespib (STA-9090) is an Hsp90 inhibitor that downregulates VEGFR, c-MET, HER2, IGF-IR, EGFR, and other Hsp90 client proteins involved in hepatocarcinogenesis, thereby making it an attractive therapy for HCC. This Phase I study was performed to establish the safety, tolerability, recommended Phase 2 dose (RP2D), and preliminary clinical activity of ganetespib in previously treated patients with advanced HCC. METHODS: Patients with advanced HCC, Child-Pugh A cirrhosis, progression on or intolerance to sorafenib, and ECOG PS ≤ 1 were enrolled in a standard 3x3 dose escalation study at doses of 100 mg/m(2), 150 mg/m(2), and 200 mg/m(2) IV given on days 1, 8, and 15 of each 28-day cycle. Objective response by RECIST version 1.1 criteria was evaluated by CT/MRI every 8 weeks. RESULTS: Fourteen patients were enrolled in this trial and received at least one dose of the study drug. Of the 14 patients: median age, 57 years old; male 71 %; Asian 36 %; HCC etiology (HBV 36 %, HCV 43 %, Hemachromatosis 7 %, unknown 21 %); Child Pugh Class (A 93 %, B 7 %); median number of prior treatments 2; median baseline AFP 70.1 ng/mL. The RP2D was determined to be 200 mg/m(2). The most commonly seen AEs were diarrhea (93 %), fatigue (71 %), AST elevation (64 %), and hyperglycemia (64 %). The most common Gr 3/4 AEs were hyperglycemia (21 %) and lipasemia (21 %). One (7 %) patient had a fatal AE, septic shock, within 30 days of receiving the study drug. One dose-limiting toxicity, grade 3 lipasemia, was observed at the 100 mg/m(2) dose. Pharmacokinetics studies showed a t1/2, CL, Tmax, and Vss of 6.45 h, 48.28 L/h (25.56 L/h/m(2)), 0.76 h, and 191 L (100.4 L/m(2)), respectively. No objective responses were seen; one patient (7 %) had stable disease at 16 weeks. Median time to progression was 1.8 months, and median overall survival was 7.2 months. CONCLUSION: Ganetespib had a manageable safety profile in patients with advanced HCC who had progressed on at least one line of systemic therapy. The pharmacokinetic profile showed that ganetespib exposure in patients with mild hepatic dysfunction is similar to that seen in patients with normal liver function. Ganetespib showed limited clinical benefit in patients with advanced HCC in this phase I trial.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Hepáticas/tratamiento farmacológico , Triazoles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Hepatocelular/metabolismo , Femenino , Humanos , Hígado/metabolismo , Hepatopatías/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Triazoles/efectos adversos , Triazoles/sangre , Triazoles/farmacocinética
9.
J Am Soc Nephrol ; 24(12): 1995-2001, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24009241

RESUMEN

Aging of the kidney is associated with renal damage, in particular mesangial matrix expansion (MME). Identifying the genes involved in this process will help to unravel the mechanisms of aging and aid in the design of novel therapeutic modalities aimed at prevention and regression. In this study, structural changes in glomeruli of 24 inbred mouse strains were characterized in male mice at 6, 12, and 20 months of age. Haplotype association mapping was used to determine genetic loci associated with the presence of MME at 20 months. This analysis identified a significant association with a 200-kb haplotype block on chromosome 6 containing Far2. Sequencing revealed that mouse strains with MME contain a 9-bp sequence in the 5' untranslated region of Far2 that is absent in most of the strains without MME. Real-time PCR showed a two-fold increase in the expression of Far2 in the kidneys of strains with the insert, and subsequent experiments performed in vitro with luciferase reporter vectors showed that this sequence difference causes differential expression of Far2. Overexpression of Far2 in a mouse mesangial cell line induced upregulation of platelet activating factor and the fibrotic marker TGF-ß. This upregulation of MME-promoting factors may result, in part, from the FAR2-catalyzed reduction of fatty acyl-coenzyme A to fatty alcohols, which are possible precursors of platelet activating factor. Overall, these data suggest the identification of a novel pathway involved in renal aging that may yield therapeutic targets for reducing MME.


Asunto(s)
Envejecimiento/patología , Aldehído Oxidorreductasas/genética , Glomérulos Renales/patología , Glomérulos Renales/fisiología , Células Mesangiales/patología , Células Mesangiales/fisiología , Envejecimiento/fisiología , Animales , Mapeo Cromosómico , Haplotipos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos MRL lpr , Ratones Endogámicos NOD , Especificidad de la Especie
10.
Geroscience ; 46(2): 2571-2581, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38103095

RESUMEN

The ability to quantify aging-related changes in histological samples is important, as it allows for evaluation of interventions intended to effect health span. We used a machine learning architecture that can be trained to detect and quantify these changes in the mouse kidney. Using additional held out data, we show validation of our model, correlation with scores given by pathologists using the Geropathology Research Network aging grading scheme, and its application in providing reproducible and quantifiable age scores for histological samples. Aging quantification also provides the insights into possible changes in image appearance that are independent of specific geropathology-specified lesions. Furthermore, we provide trained classifiers for H&E-stained slides, as well as tutorials on how to use these and how to create additional classifiers for other histological stains and tissues using our architecture. This architecture and combined resources allow for the high throughput quantification of mouse aging studies in general and specifically applicable to kidney tissues.


Asunto(s)
Envejecimiento , Aprendizaje Automático , Ratones , Animales , Envejecimiento/patología , Riñón
11.
Biomolecules ; 14(5)2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38786006

RESUMEN

Age is the primary risk factor for neurodegenerative diseases such as Alzheimer's and Huntington's disease. Alzheimer's disease is the most common form of dementia and a leading cause of death in the elderly population of the United States. No effective treatments for these diseases currently exist. Identifying effective treatments for Alzheimer's, Huntington's, and other neurodegenerative diseases is a major current focus of national scientific resources, and there is a critical need for novel therapeutic strategies. Here, we investigate the potential for targeting the kynurenine pathway metabolite 3-hydroxyanthranilic acid (3HAA) using Caenorhabditis elegans expressing amyloid-beta or a polyglutamine peptide in body wall muscle, modeling the proteotoxicity in Alzheimer's and Huntington's disease, respectively. We show that knocking down the enzyme that degrades 3HAA, 3HAA dioxygenase (HAAO), delays the age-associated paralysis in both models. This effect on paralysis was independent of the protein aggregation in the polyglutamine model. We also show that the mechanism of protection against proteotoxicity from HAAO knockdown is mimicked by 3HAA supplementation, supporting elevated 3HAA as the mediating event linking HAAO knockdown to delayed paralysis. This work demonstrates the potential for 3HAA as a targeted therapeutic in neurodegenerative disease, though the mechanism is yet to be explored.


Asunto(s)
Ácido 3-Hidroxiantranílico , Péptidos beta-Amiloides , Caenorhabditis elegans , Parálisis , Péptidos , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Animales , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/genética , Péptidos/farmacología , Ácido 3-Hidroxiantranílico/metabolismo , Parálisis/inducido químicamente , Parálisis/metabolismo , Parálisis/genética , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/genética , Dioxigenasas/metabolismo , Dioxigenasas/genética
12.
bioRxiv ; 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37461572

RESUMEN

The ability to quantify aging-related changes in histological samples is important, as it allows for evaluation of interventions intended to effect health span. We used a machine learning architecture that can be trained to detect and quantify these changes in the mouse kidney. Using additional held out data, we show validation of our model, correlation with scores given by pathologists using the Geropathology Research Network aging grading scheme, and its application in providing reproducible and quantifiable age scores for histological samples. Aging quantification also provides the insights into possible changes in image appearance that are independent of specific geropathology-specified lesions. Furthermore, we provide trained classifiers for H&E-stained slides, as well as tutorials on how to use these and how to create additional classifiers for other histological stains and tissues using our architecture.This architecture and combined resources allow for the high throughput quantification of mouse aging studies in general and specifically applicable to kidney tissues.

13.
Nat Commun ; 14(1): 8338, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38097593

RESUMEN

Tryptophan metabolism through the kynurenine pathway influences molecular processes critical to healthy aging including immune signaling, redox homeostasis, and energy production. Aberrant kynurenine metabolism occurs during normal aging and is implicated in many age-associated pathologies including chronic inflammation, atherosclerosis, neurodegeneration, and cancer. We and others previously identified three kynurenine pathway genes-tdo-2, kynu-1, and acsd-1-for which decreasing expression extends lifespan in invertebrates. Here we report that knockdown of haao-1, a fourth gene encoding the enzyme 3-hydroxyanthranilic acid (3HAA) dioxygenase (HAAO), extends lifespan by ~30% and delays age-associated health decline in Caenorhabditis elegans. Lifespan extension is mediated by increased physiological levels of the HAAO substrate 3HAA. 3HAA increases oxidative stress resistance and activates the Nrf2/SKN-1 oxidative stress response. In pilot studies, female Haao knockout mice or aging wild type male mice fed 3HAA supplemented diet were also long-lived. HAAO and 3HAA represent potential therapeutic targets for aging and age-associated disease.


Asunto(s)
Proteínas de Caenorhabditis elegans , Quinurenina , Animales , Masculino , Femenino , Ratones , Quinurenina/metabolismo , Triptófano/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Ácido 3-Hidroxiantranílico/metabolismo , Longevidad/genética , Ratones Noqueados , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo
14.
Am J Physiol Renal Physiol ; 303(7): F972-81, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22859403

RESUMEN

Albuminuria is an important marker of nephropathy that increases the risk of progressive renal and chronic cardiovascular diseases. The genetic basis of kidney disease is well-established in humans and rodent models, but the causal genes remain to be identified. We applied several genetic strategies to map and refine genetic loci affecting albuminuria in mice and translated the findings to human kidney disease. First, we measured albuminuria in mice from 33 inbred strains, used the data for haplotype association mapping (HAM), and detected 10 genomic regions associated with albuminuria. Second, we performed eight F(2) intercrosses between genetically diverse strains to identify six loci underlying albuminuria, each of which was concordant to kidney disease loci in humans. Third, we used the Oak Ridge National Laboratory incipient Collaborative Cross subpopulation to detect an additional novel quantitative trait loci (QTL) underlying albuminuria. We also performed a ninth intercross, between genetically similar strains, that substantially narrowed an albuminuria QTL on Chromosome 17 to a region containing four known genes. Finally, we measured renal gene expression in inbred mice to detect pathways highly correlated with albuminuria. Expression analysis also identified Glcci1, a gene known to affect podocyte structure and function in zebrafish, as a strong candidate gene for the albuminuria QTL on Chromosome 6. Overall, these findings greatly enhance our understanding of the genetic basis of albuminuria in mice and may guide future studies into the genetic basis of kidney disease in humans.


Asunto(s)
Albuminuria/genética , Riñón/metabolismo , Sitios de Carácter Cuantitativo , Albuminuria/metabolismo , Animales , Cruzamientos Genéticos , Expresión Génica , Estudios de Asociación Genética , Haplotipos , Ratones , Ratones Endogámicos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
15.
Bioorg Med Chem Lett ; 22(22): 6974-9, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23046966

RESUMEN

AZD9272 and AZD6538 are two novel mGluR5 negative allosteric modulators selected for further clinical development. An initial high-throughput screening revealed leads with promising profiles, which were further optimized by minor, yet indispensable, structural modifications to bring forth these drug candidates. Advantageously, both compounds may be synthesized in as little as one step. Both are highly potent and selective for the human as well as the rat mGluR5 where they interact at the same binding site than MPEP. They are orally available, allow for long interval administration due to a high metabolic stability and long half-lives in rats and permeate the blood brain barrier to a high extent. AZD9272 has progressed into phase I clinical studies.


Asunto(s)
Oxadiazoles/química , Piridinas/química , Receptores de Glutamato Metabotrópico/química , Regulación Alostérica , Animales , Sitios de Unión , Sistema Nervioso Central/diagnóstico por imagen , Evaluación Preclínica de Medicamentos , Células HEK293 , Semivida , Humanos , Marcaje Isotópico , Masculino , Microsomas/metabolismo , Oxadiazoles/síntesis química , Oxadiazoles/farmacocinética , Piridinas/síntesis química , Piridinas/farmacocinética , Cintigrafía , Ratas , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/metabolismo , Relación Estructura-Actividad
16.
Hosp Top ; 100(2): 69-76, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34470597

RESUMEN

The 2019 SARS-CoV2 virus presented a capacity demand scenario for Yale New Haven Hospital. The response was created with a focus on clinical needs, but was also driven by the unique characteristics of the buildings within our institution. These physical characteristics were considered in the response as a safety measure as little was known about the transmissibility risk in the acute hospital setting of SARS-CoV2 at the time of response. The lessons learned in capacity expansion to meet the potentially catastrophic demand for acute care services due to a novel, poorly understood pathogen are discussed here.


Asunto(s)
COVID-19 , Hospitales , Humanos , Pacientes Internos , Pandemias/prevención & control , ARN Viral , SARS-CoV-2
17.
J Lipid Res ; 52(6): 1139-1149, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21444760

RESUMEN

Genome-wide association (GWA) studies represent a powerful strategy for identifying susceptibility genes for complex diseases in human populations but results must be confirmed and replicated. Because of the close homology between mouse and human genomes, the mouse can be used to add evidence to genes suggested by human studies. We used the mouse quantitative trait loci (QTL) map to interpret results from a GWA study for genes associated with plasma HDL cholesterol levels. We first positioned single nucleotide polymorphisms (SNPs) from a human GWA study on the genomic map for mouse HDL QTL. We then used mouse bioinformatics, sequencing, and expression studies to add evidence for one well-known HDL gene (Abca1) and three newly identified genes (Galnt2, Wwox, and Cdh13), thus supporting the results of the human study. For GWA peaks that occur in human haplotype blocks with multiple genes, we examined the homologous regions in the mouse to prioritize the genes using expression, sequencing, and bioinformatics from the mouse model, showing that some genes were unlikely candidates and adding evidence for candidate genes Mvk and Mmab in one haplotype block and Fads1 and Fads2 in the second haplotype block. Our study highlights the value of mouse genetics for evaluating genes found in human GWA studies.


Asunto(s)
HDL-Colesterol/genética , Genoma Humano , Genoma , Ratones/genética , Sitios de Carácter Cuantitativo , Secuencia de Aminoácidos , Animales , HDL-Colesterol/sangre , Mapeo Cromosómico , Biología Computacional , delta-5 Desaturasa de Ácido Graso , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Ratones Endogámicos , Datos de Secuencia Molecular , Homología de Secuencia
18.
Lancet Oncol ; 11(1): 48-54, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19932054

RESUMEN

BACKGROUND: Previous phase 2 studies have shown antitumour activity with gemcitabine and oxaliplatin (GEMOX) in patients with advanced biliary-tract cancers (BTCs). In this phase 2 study, we assessed the efficacy and safety of combined bevacizumab with GEMOX (GEMOX-B) in patients with advanced BTCs, and investigated how changes in 18-fluorodeoxyglucose ([(18)F]FDG)-PET correlate with clinical outcome. METHODS: Patients with advanced measurable BTCs were given the following treatment on days 1 and 15 of a 28-day cycle: bevacizumab 10 mg/kg, followed by gemcitabine 1000 mg/m(2) (10 mg/m(2) per min) and oxaliplatin 85 mg/m(2) (2-h infusion). [(18)F]FDG-PET scans were obtained at baseline and after completion of the second cycle. The primary endpoint was progression-free survival (PFS). Efficacy and safety analyses were by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00361231. FINDINGS: 35 patients were enrolled and evaluable for efficacy and toxicity. Median PFS was 7.0 months (95% CI 5.3-10.3), and PFS at 6 months was 63% (47-79), which was below the targeted rate of 70%. Grade 3-4 toxic effects included neutropenia (n=7), raised alanine aminotransferase concentrations (n=5), peripheral neuropathy (n=5), and hypertension (n=5). [(18)F]FDG-PET scans showed a significant decrease in maximum standardised uptake value (SUV(max)) after two cycles of treatment (5.72 [SD 2.01] at baseline; 3.73 [SD 1.88] after two cycles; p<0.0001). These changes were more pronounced in patients with partial response or stable disease than those with progressive disease (24 patients, -2.80 [SD 1.95] vs five patients, 1.41 [SD 3.13]; p=0.009). Change in SUV(max) was a significant predictor of PFS (HR 1.35, 1.14-1.60, p=0.0006) and overall survival (1.25, 1.05-1.50, p=0.01). INTERPRETATION: GEMOX-B showed antitumour activity with tolerable safety in patients with advanced BTCs. Decreases in SUV(max) on [(18)F]FDG-PET scans after treatment were associated with disease control and increases in PFS and overall survival. FUNDING: Genentech Oncology and Sanofi-Aventis.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Sistema Biliar/diagnóstico por imagen , Neoplasias del Sistema Biliar/tratamiento farmacológico , Fluorodesoxiglucosa F18 , Tomografía de Emisión de Positrones , Radiofármacos , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bevacizumab , Neoplasias del Sistema Biliar/mortalidad , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/uso terapéutico , Modelos de Riesgos Proporcionales , Medición de Riesgo , Factores de Tiempo , Resultado del Tratamiento
19.
Bioorg Med Chem Lett ; 20(17): 5184-90, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20656488

RESUMEN

Succinic acid amides have been found to be effective P2-P3 scaffold replacements for peptidic ICE inhibitors. Heteroarylalkyl fragments occupying the P4 position provided access to compounds with nM affinities. Utilization of an acylal prodrug moiety was required to overcome biopharmaceutical issues which led to the identification of 17f, a potential clinical candidate.


Asunto(s)
Amidas/química , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/farmacología , Ácido Succínico/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacocinética , Semivida , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Estereoisomerismo , Relación Estructura-Actividad
20.
Genetics ; 179(1): 693-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18493081

RESUMEN

We investigated the effect of apolipoprotein E (Apoe) on albuminuria in the males of two independent F2 intercrosses between C57BL/6J and A/J mice, using wild-type inbred strains in the first cross and B6-Apoe(-/-) animals in the second cross. In the first cross, we identified three quantitative trait loci (QTL): chromosome (Chr) 2 [LOD 3.5, peak at 70 cM, confidence interval (C.I.) 28-88 cM]; Chr 9 (LOD 2.0, peak 5 cM, C.I. 5-25 cM); and Chr 19 (LOD 1.9, peak 49 cM, C.I. 23-54 cM). The Chr 2 and Chr 19 QTL were concordant with previously found QTL for renal damage in rat and human. The Chr 9 QTL was concordant with a locus found in rat. The second cross, testing only Apoe(-/-) progeny, did not identify any of these loci, but detected two other loci on Chr 4 (LOD 3.2, peak 54 cM, C.I. 29-73 cM) and Chr 6 (LOD 2.6, peak 33 cM, C.I. 11-61 cM), one of which was concordant with a QTL found in rat. The dependence of QTL detection on the presence of Apoe and the concordance of these QTL with rat and human kidney disease QTL suggest that Apoe plays a role in renal damage.


Asunto(s)
Albuminuria/genética , Apolipoproteínas E/genética , Sitios de Carácter Cuantitativo/genética , Animales , Cruzamientos Genéticos , Genotipo , Escala de Lod , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA