Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lab Invest ; 104(1): 100282, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37924947

RESUMO

Large-scale high-dimensional multiomics studies are essential to unravel molecular complexity in health and disease. We developed an integrated system for tissue sampling (CryoGrid), analytes preparation (PIXUL), and downstream multiomic analysis in a 96-well plate format (Matrix), MultiomicsTracks96, which we used to interrogate matched frozen and formalin-fixed paraffin-embedded (FFPE) mouse organs. Using this system, we generated 8-dimensional omics data sets encompassing 4 molecular layers of intracellular organization: epigenome (H3K27Ac, H3K4m3, RNA polymerase II, and 5mC levels), transcriptome (messenger RNA levels), epitranscriptome (m6A levels), and proteome (protein levels) in brain, heart, kidney, and liver. There was a high correlation between data from matched frozen and FFPE organs. The Segway genome segmentation algorithm applied to epigenomic profiles confirmed known organ-specific superenhancers in both FFPE and frozen samples. Linear regression analysis showed that proteomic profiles, known to be poorly correlated with transcriptomic data, can be more accurately predicted by the full suite of multiomics data, compared with using epigenomic, transcriptomic, or epitranscriptomic measurements individually.


Assuntos
Formaldeído , Proteômica , Camundongos , Animais , Fixadores , Fixação de Tecidos/métodos , Proteômica/métodos , Inclusão em Parafina/métodos
2.
bioRxiv ; 2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-36993219

RESUMO

Background: The multiome is an integrated assembly of distinct classes of molecules and molecular properties, or "omes," measured in the same biospecimen. Freezing and formalin-fixed paraffin-embedding (FFPE) are two common ways to store tissues, and these practices have generated vast biospecimen repositories. However, these biospecimens have been underutilized for multi-omic analysis due to the low throughput of current analytical technologies that impede large-scale studies. Methods: Tissue sampling, preparation, and downstream analysis were integrated into a 96-well format multi-omics workflow, MultiomicsTracks96. Frozen mouse organs were sampled using the CryoGrid system, and matched FFPE samples were processed using a microtome. The 96-well format sonicator, PIXUL, was adapted to extract DNA, RNA, chromatin, and protein from tissues. The 96-well format analytical platform, Matrix, was used for chromatin immunoprecipitation (ChIP), methylated DNA immunoprecipitation (MeDIP), methylated RNA immunoprecipitation (MeRIP), and RNA reverse transcription (RT) assays followed by qPCR and sequencing. LC-MS/MS was used for protein analysis. The Segway genome segmentation algorithm was used to identify functional genomic regions, and linear regressors based on the multi-omics data were trained to predict protein expression. Results: MultiomicsTracks96 was used to generate 8-dimensional datasets including RNA-seq measurements of mRNA expression; MeRIP-seq measurements of m6A and m5C; ChIP-seq measurements of H3K27Ac, H3K4m3, and Pol II; MeDIP-seq measurements of 5mC; and LC-MS/MS measurements of proteins. We observed high correlation between data from matched frozen and FFPE organs. The Segway genome segmentation algorithm applied to epigenomic profiles (ChIP-seq: H3K27Ac, H3K4m3, Pol II; MeDIP-seq: 5mC) was able to recapitulate and predict organ-specific super-enhancers in both FFPE and frozen samples. Linear regression analysis showed that proteomic expression profiles can be more accurately predicted by the full suite of multi-omics data, compared to using epigenomic, transcriptomic, or epitranscriptomic measurements individually. Conclusions: The MultiomicsTracks96 workflow is well suited for high dimensional multi-omics studies - for instance, multiorgan animal models of disease, drug toxicities, environmental exposure, and aging as well as large-scale clinical investigations involving the use of biospecimens from existing tissue repositories.

3.
Sci Rep ; 10(1): 10397, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32587356

RESUMO

Serum amyloid A3 (Saa3) derives mainly from extrahepatic tissue and is not detected in plasma from moderately inflamed obese mice. In contrast, it is present in plasma from mice acutely inflamed by injection of high dose of lipopolysaccharide (LPS). To reconcile these differences, we evaluated whether different acute inflammatory stimuli could affect the presence of Saa3 in plasma. Saa3 appeared dose dependently in plasma after LPS injection. In contrast, only very low levels were detected after sterile inflammation with silver nitrate despite levels of Saa1 and Saa2 being comparable to high dose LPS. Saa3 was not detected in plasma following casein administration. Although most Saa3 was found in HDL, a small amount was not lipoprotein associated. Gene expression and proteomic analysis of liver and adipose tissue suggested that a major source of Saa3 in plasma after injection of LPS was adipose tissue rather than liver. We conclude that Saa3 only appears in plasma after induction of acute inflammation by some but not all inflammatory stimuli. These findings are consistent with the observation that Saa3 is not detectable in plasma in more moderate chronic inflammatory states such as obesity.


Assuntos
Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Proteína Amiloide A Sérica/fisiologia , Nitrato de Prata/toxicidade , Animais , Anti-Infecciosos Locais/farmacologia , Anti-Infecciosos Locais/toxicidade , Inflamação/sangue , Inflamação/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
J Clin Lipidol ; 14(1): 66-76.e11, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31859127

RESUMO

BACKGROUND: Humans spend most of the time in the postprandial state, yet most knowledge about high-density lipoproteins (HDL) derives from the fasted state. HDL protein and lipid cargo mediate HDL's antiatherogenic effects, but whether these HDL constituents change in the postprandial state and are affected by dietary macronutrients remains unknown. OBJECTIVES: This study aimed to assess changes in HDL protein and lipid composition after the consumption of a high-carbohydrate or high saturated fat (HSF) meal. METHODS: We isolated HDL from plasma collected during a randomized, cross-over study of metabolically healthy subjects. Subjects consumed isocaloric meals consisting predominantly of either carbohydrate or fat. At baseline and at 3 and 6 hours postprandial, we quantified HDL protein and lipid composition by liquid chromatography-mass spectrometry. RESULTS: A total of 15 subjects were included (60% female, aged 34 ± 15 years, body mass index: 24.1 ± 2.7 kg/m2). Consumption of the HSF meal led to HDL enrichment in total lipid (P = .006), triglyceride (P = .02), and phospholipid (P = .008) content and a corresponding depletion in protein content. After the HSF meal, 16 of the 25 measured phosphatidylcholine species significantly increased in abundance (P values range from .027 to <.001), along with several sphingolipids including ceramides (P < .004), lactosylceramide (P = .023), and sphingomyelin-14 (P = .013). Enrichment in apolipoprotein A-I (P = .001) was the only significant change in HDL protein composition after the HSF meal. The high-carbohydrate meal conferred only minimal changes in HDL composition. CONCLUSION: Meal macronutrient content acutely affects HDL composition in the postprandial state, with the HSF meal resulting in enrichment of HDL phospholipid content with possible consequences for HDL function.


Assuntos
Carboidratos/administração & dosagem , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/sangue , Lipoproteínas HDL/sangue , Obesidade/sangue , Adulto , Glicemia/genética , Índice de Massa Corporal , Carboidratos/efeitos adversos , LDL-Colesterol/sangue , Gorduras na Dieta/efeitos adversos , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Jejum , Feminino , Humanos , Lipidômica/métodos , Masculino , Refeições , Obesidade/dietoterapia , Obesidade/genética , Obesidade/patologia , Período Pós-Prandial/genética , Triglicerídeos/sangue
6.
Diabetes Care ; 41(11): 2361-2369, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30150236

RESUMO

OBJECTIVE: We examined the association of urine complement proteins with progression to end-stage renal disease (ESRD) or death in people with type 2 diabetes and proteinuric diabetic kidney disease (DKD). RESEARCH DESIGN AND METHODS: Using targeted mass spectrometry, we quantified urinary abundance of 12 complement proteins in a predominantly Mexican American cohort with type 2 diabetes and proteinuric DKD (n = 141). The association of urine complement proteins with progression to ESRD or death was evaluated using time-to-event analyses. RESULTS: At baseline, median estimated glomerular filtration rate (eGFR) was 54 mL/min/1.73 m2 and urine protein-to-creatinine ratio 2.6 g/g. Sixty-seven participants developed ESRD or died, of whom 39 progressed to ESRD over a median of 3.1 years and 40 died over a median 3.6 years. Higher urine CD59, an inhibitor of terminal complement complex formation, was associated with a lower risk of ESRD (hazard ratio [HR] [95% CI per doubling] 0.50 [0.29-0.87]) and death (HR [95% CI] 0.56 [0.34-0.93]), after adjustment for demographic and clinical covariates, including baseline eGFR and proteinuria. Higher urine complement components 4 and 8 were associated with lower risk of death (HR [95% CI] 0.57 [0.41-0.79] and 0.66 [0.44-0.97], respectively); higher urine factor H-related protein 2, a positive regulator of the alternative complement pathway, was associated with greater risk of death (HR [95% CI] 1.61 [1.05-2.48]) in fully adjusted models. CONCLUSIONS: In a largely Mexican American cohort with type 2 diabetes and proteinuric DKD, urine abundance of several complement and complement regulatory proteins was strongly associated with progression to ESRD and death.


Assuntos
Proteínas do Sistema Complemento/urina , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/mortalidade , Diabetes Mellitus Tipo 2/urina , Nefropatias Diabéticas/diagnóstico , Falência Renal Crônica/diagnóstico , Idoso , Estudos Transversais , Diabetes Mellitus Tipo 2/etnologia , Nefropatias Diabéticas/etnologia , Nefropatias Diabéticas/mortalidade , Nefropatias Diabéticas/urina , Progressão da Doença , Feminino , Taxa de Filtração Glomerular , Humanos , Falência Renal Crônica/etnologia , Falência Renal Crônica/mortalidade , Falência Renal Crônica/urina , Masculino , Espectrometria de Massas , Americanos Mexicanos , Pessoa de Meia-Idade , Prognóstico , Proteinúria/diagnóstico , Proteinúria/etnologia , Proteinúria/mortalidade , Proteinúria/patologia , Fatores de Risco , Análise de Sobrevida
7.
Cell Rep ; 23(10): 3021-3030, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29874587

RESUMO

Type 2 diabetes (T2D) is associated with increased risk for atherosclerosis; however, the mechanisms underlying this relationship are poorly understood. Macrophages, which are activated in T2D and causatively linked to atherogenesis, are an attractive mechanistic link. Here, we use proteomics to show that diet-induced obesity and insulin resistance (obesity/IR) modulate a pro-atherogenic "macrophage-sterol-responsive-network" (MSRN), which, in turn, predisposes macrophages to cholesterol accumulation. We identify IFNγ as the mediator of obesity/IR-induced MSRN dysregulation and increased macrophage cholesterol accumulation and show that obesity/IR primes T cells to increase IFNγ production. Accordingly, myeloid cell-specific deletion of the IFNγ receptor (Ifngr1-/-) restores MSRN proteins, attenuates macrophage cholesterol accumulation and atherogenesis, and uncouples the strong relationship between hyperinsulinemia and aortic root lesion size in hypercholesterolemic Ldlr-/- mice with obesity/IR, but does not affect these parameters in Ldlr-/- mice without obesity/IR. Collectively, our findings identify an IFNγ-macrophage pathway as a mechanistic link between obesity/IR and accelerated atherogenesis.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Resistência à Insulina , Interferon gama/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Animais , Aterosclerose/genética , Colesterol/metabolismo , Células Espumosas/metabolismo , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Receptores de Interferon/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/metabolismo , Receptor de Interferon gama
8.
J Lipid Res ; 56(8): 1519-30, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25995210

RESUMO

Recent studies demonstrate that HDL's ability to promote cholesterol efflux from macrophages associates strongly with cardioprotection in humans independently of HDL-cholesterol (HDL-C) and apoA-I, HDL's major protein. However, the mechanisms that impair cholesterol efflux capacity during vascular disease are unclear. Inflammation, a well-established risk factor for cardiovascular disease, has been shown to impair HDL's cholesterol efflux capacity. We therefore tested the hypothesis that HDL's impaired efflux capacity is mediated by specific changes of its protein cargo. Humans with acute inflammation induced by low-level endotoxin had unchanged HDL-C levels, but their HDL-C efflux capacity was significantly impaired. Proteomic analyses demonstrated that HDL's cholesterol efflux capacity correlated inversely with HDL content of serum amyloid A (SAA)1 and SAA2. In mice, acute inflammation caused a marked impairment of HDL-C efflux capacity that correlated with a large increase in HDL SAA. In striking contrast, the efflux capacity of mouse inflammatory HDL was preserved with genetic ablation of SAA1 and SAA2. Our observations indicate that the inflammatory impairment of HDL-C efflux capacity is due in part to SAA-mediated remodeling of HDL's protein cargo.


Assuntos
HDL-Colesterol/metabolismo , Proteoma/metabolismo , Adulto , Animais , HDL-Colesterol/sangue , HDL-Colesterol/química , Citoproteção , Endotoxinas/toxicidade , Humanos , Inflamação/sangue , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Miocárdio/citologia , Miocárdio/metabolismo , Proteína Amiloide A Sérica/deficiência , Proteína Amiloide A Sérica/metabolismo
9.
Semin Oncol ; 32(6 Suppl 9): S11-5, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16399423

RESUMO

The dramatically increased incidence and poor survival rates of esophageal adenocarcinoma (EAC) underscore the need for novel targets useful for risk assessment and therapeutic intervention. Altered expression of cyclin D1 has been proposed as an early predictor for malignant transformation in EAC; however, the mechanisms underlying cyclin D1 deregulation have not been identified. A single nucleotide polymorphism, A870G, of the cyclin D1 gene has been associated with the preferential encoding of a protein with an extended half-life. We investigated the association of the cyclin D1 A870G polymorphism with cyclin D1 protein expression and clinical characteristics and outcome in 124 patients treated at our institution for EAC. Our results indicate that the cyclin D1 AA/AG genotype is associated with earlier age of cancer onset, cyclin D1 protein deregulation in the primary tumors, and increased frequency of distant metastasis. Our findings suggest that cyclin D1 status could be useful to assess risk of progression to EAC, and strategies directed to modulate cyclin D1 expression may prove useful for interventions to slow or interrupt the EAC tumorigenesis process.


Assuntos
Adenocarcinoma/genética , Ciclina D1/genética , Neoplasias Esofágicas/genética , Polimorfismo Genético , Adenocarcinoma/patologia , Biomarcadores Tumorais/genética , Neoplasias Esofágicas/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Estudos Retrospectivos
10.
J Natl Cancer Inst ; 95(3): 198-205, 2003 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-12569141

RESUMO

BACKGROUND: Altered cyclin D1 expression in advanced preinvasive lesions of the upper aerodigestive tract (UADT) is associated with an increased risk of developing cancer and histologic progression during and after combination biochemopreventive therapy (13-cis-retinoic acid, alpha-interferon, and alpha-tocopherol). Both alleles of the adenine (A)/guanine (G) cyclin D1 polymorphism located at nucleotide 870 encode two alternatively spliced transcripts, but the A allele preferentially encodes a protein with an extended half-life. We investigated whether the cyclin D1 genotype at nucleotide 870 was associated with baseline levels of cyclin D1 protein, post-treatment modulation of cyclin D1 protein levels, histologic response to treatment, and the outcome for subjects with preinvasive UADT lesions after biochemopreventive therapy. METHODS: UADT tissue biopsy samples were obtained before and 6 and 12 months after biochemopreventive treatment from 31 individuals with advanced preinvasive UADT lesions. Tissues were examined for cyclin D1 genotype (by DNA single-strand conformation polymorphism analysis), for cyclin D1 protein expression (by immunohistochemistry), and for cyclin D1 gene copy number (by fluorescence in situ hybridization). Associations of cyclin D1 genotype with histologic response to therapy and time to progression to a higher degree of dysplasia or invasive cancer were investigated. All statistical tests were two-sided. RESULTS: The A allele was associated with increased baseline cyclin D1 expression in the parabasal epithelial layer (16 of 18 AA/AG subjects versus four of nine GG subjects; P =.02), decreased histologic response to biochemopreventive treatment (six of 21 AA/AG subjects versus four of 10 GG subjects; P =.70), decreased favorable modulation of cyclin D1 expression by the treatment (seven of 18 AA/AG subjects versus eight of nine GG subjects; P =.02), and shorter progression-free survival (P =.05). CONCLUSIONS: The cyclin D1 A allele was associated with a diminished modulation of normal physiologic and treatment-induced decreased expression of cyclin D1, a decreased likelihood of response to biochemopreventive intervention, and an increased rate of progression to cancer development, findings that require validation in a larger cohort.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclina D1/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/prevenção & controle , Neoplasias Bucais/genética , Neoplasias Bucais/prevenção & controle , Ciclina D1/sangue , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Genótipo , Humanos , Imuno-Histoquímica , Fatores Imunológicos/uso terapêutico , Hibridização in Situ Fluorescente , Interferon-alfa/administração & dosagem , Isotretinoína/administração & dosagem , Neoplasias Laríngeas/sangue , Neoplasias Laríngeas/patologia , Neoplasias Bucais/sangue , Neoplasias Bucais/patologia , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Estudos Prospectivos , Resultado do Tratamento , alfa-Tocoferol/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...