Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Anim Nutr ; 17: 232-243, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38800739

RESUMEN

This study aimed to understand how molecular mechanisms controlling water and urea metabolism at the finishing phase can be affected by previous plane of nutrition of crossbred Angus beef steers. Twenty-four (n = 24) animals were randomly distributed into either a moderate (MP) or high plane of nutrition during the background phase for 85 d. Animals were then blocked by their previous plane and were moved onto a 105-d finishing phase in a 2 × 2 factorial arrangement. The forage-finished group received only high-quality alfalfa hay, whereas the grain-fed group received a high grain diet (80% whole corn and 20% alfalfa hay). By the end of the finishing phase, animals were harvested and tissue samples from the rumen and kidney were collected. Changes in gene expression of aquaporins (AQP)-2, -3, -4, -7, ATP1A1, ATP1B1, SGK1, CLIC1 (kidney and rumen), UT-A1 (kidney only) and UT-B (rumen only), were assayed via real-time qPCR; 18S rRNA was used as an endogenous control. One-way ANOVA followed by Tukey's post hoc analysis was conducted. When animals were from MP, forage-finishing increased the relative abundance of AQP3 (P ≤ 0.05), AQP7 (P ≤ 0.05), ATP1B1 (P ≤ 0.05), and SGK1 (P ≤ 0.05) in the kidney when compared to grain-fed animals. In the rumen, for the MP group, AQP7 was differentially expressed in both treatments at the finishing phase (P ≤ 0.01), with forage-finished steers having the highest expression of AQP7. For the MP group, UT-B had a tendency of presenting a higher expression on grain-fed animals (P = 0.075). Overall, these results suggest that previous plane can impact expression of genes associated with water and urea metabolism during the finishing phase, namely AQP3, AQP7, ATP1B1, and SGK1 in the kidney, and AQP7 and UT-B in the rumen. The greatest impact observed on gene expression changes of investigated genes at the finishing phase was reflective of animals backgrounded on the moderate previous plane.

2.
Biomolecules ; 13(9)2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37759673

RESUMEN

OBJECTIVE: Evidence suggests that food bioactives affect the epigenome to prevent pathological cardiac hypertrophy. Recently, we showed that emodin, an anthraquinone, attenuated pathological cardiac hypertrophy and histone deacetylase (HDAC) activity. However, we only examined the cardioprotective effects of emodin's parent compound and not those of emodin metabolites or of emodin-gut microbiome interactions. The microbiome has emerged as a key player in chronic diseases such as metabolic and cardiac disease. Thus, we hypothesized that emodin could reverse hypertension-induced changes in microbial communities. METHODS: Normo- and hypertensive (angiotensin II) C57/BL6 female mice were randomly assigned to receive a vehicle (Veh; DMSO:PEG 1:1) or emodin (Emod; 30 mg/kg) for 14 days. Body weights were collected pre- and post-treatment, and blood pressure was assessed via tail cuff. At the study's end, the mice were euthanized and assessed for their heart weights. In addition, stool samples and cecal contents were collected to elucidate changes in the microbial populations using 16S rRNA sequencing. Lastly, the tissue was lysed, and RNA was isolated for qPCR. One-way ANOVA with Tukey's post hoc test was performed unless otherwise specified, and p < 0.05 was considered significant. RESULTS: Emodin significantly attenuated cardiac hypertrophy in the female mice. No significant changes were observed in body weight or systolic blood pressure in response to hypertension or emodin. Lastly, analysis suggests that hypertension altered the microbiome in the cecum and cecal content, with additional evidence to support that emodin affects gut microbiota in the feces and colon. CONCLUSIONS: Our data demonstrate that emodin attenuates pathological hypertrophy in female mice. Future research is needed to dissect if changes in the microbiome contributes to emodin-mediated attenuation in cardiac remodeling.


Asunto(s)
Emodina , Microbioma Gastrointestinal , Hipertensión , Animales , Femenino , Ratones , Angiotensinas/toxicidad , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Emodina/farmacología , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/patología , ARN Ribosómico 16S/genética
3.
Cell Signal ; 110: 110815, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37478958

RESUMEN

Skeletal muscle atrophy is defined by wasting or decrease in muscle mass owing to injury, aging, malnutrition, chronic disuse, or physical consequences of chronic illness. Under normal physiological conditions, a network of signal transduction pathways serves to balance muscle protein synthesis and proteolysis; however, metabolic shifts occur from protein synthesis to protein degradation that leads to a reduction in cross-sectional myofibers and can result in loss of skeletal muscle mass (atrophy) over time. Recent evidence highlights posttranslational modifications (PTMs) such as acetylation and phosphorylation in contractile dysfunction and muscle wasting. Indeed, histone deacetylase (HDAC) inhibitors have been shown to attenuate muscle atrophy and delay muscle damage in response to nutrient deprivation, in models of metabolic dysfunction and genetic models of muscle disease (e.g., muscle dystrophy). Despite our current understanding of lysine acetylation in muscle physiology, a role for HDACs in the regulation of muscle signal transduction remains a 'black box.' Using C2C12 myotubes stimulated with dexamethasone (Dex) as a model of muscle atrophy, we report that protein kinase C delta (PKCδ) phosphorylation decreased at threonine 505 (T505) and serine 643 (S643) in myotubes in response to muscle atrophy; these residues are important for PKCδ activity. Interestingly, PKCδ phosphorylation was restored/increased in myotubes treated with a pan-HDAC inhibitor or a class I selective HDAC inhibitor targeting HDACs1, -2, and - 3 in response to Dex. Moreover, we observed that Dex induced atrophy in skeletal muscle tissue in mice; this reduction in atrophy occurred rapidly, with weight loss noted by day 3 post-Dex and muscle weight loss noted by day 7. Similar to our findings in C2C12 myotubes, Dex attenuated phosphorylation of PKCδ at S643, while HDAC inhibition restored or increased PKCδ phosphorylation at both T505 and S643 in the tibialis anterior. Consistent with this hypothesis, we report that HDAC inhibition could not restore myotube size in response to Dex in the presence of a PKCδ inhibitor or when overexpressing a dominant negative PKCδ. Additionally, the overexpression of a constitutively active PKCδ prevented Dex-induced myotube atrophy. Combined, these data suggest that HDACs regulate muscle physiology via changes in intracellular signaling, namely PKCδ phosphorylation. Whether HDACs regulate PKCδ through canonical (e.g. gene-mediated regulation of phosphatases) or non-canonical (e.g. direct deacetylation of PKCδ to change phosphorylation states) mechanisms remain unclear and future research is needed to clarify this point.


Asunto(s)
Inhibidores de Histona Desacetilasas , Proteína Quinasa C-delta , Ratones , Animales , Inhibidores de Histona Desacetilasas/farmacología , Fosforilación , Proteína Quinasa C-delta/metabolismo , Estudios Transversales , Atrofia Muscular/metabolismo , Músculo Esquelético/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Dexametasona/efectos adversos , Dexametasona/metabolismo , Pérdida de Peso
4.
Biochem Biophys Res Commun ; 650: 81-86, 2023 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-36773343

RESUMEN

Skeletal muscle differentiation involves activation of quiescent satellite cells to proliferate, differentiate and fuse to form new myofibers; this requires coordination of myogenic transcription factors. Myogenic transcription is tightly regulated by various intracellular signaling pathways, which include members of the protein kinase D (PKD) family. PKD is a family of serine-threonine kinases that regulate gene expression, protein secretion, cell proliferation, differentiation and inflammation. PKD is a unique PKC family member that shares distant sequence homology to calcium-regulated kinases and plays an important role in muscle physiology. In this report, we show that class I histone deacetylase (HDAC) inhibition, and in particular HDAC8 inhibition, attenuated PKD phosphorylation in skeletal C2C12 myoblasts in response to phorbol ester, angiotensin II and dexamethasone signaling independent of changes in total PKD protein expression. As class I HDACs and PKD signaling are requisite for myocyte differentiation, these data suggest that HDAC8 functions as a potential feedback regulator of PKD phosphorylation to control myogenic gene expression.


Asunto(s)
Mioblastos Esqueléticos , Proteína Quinasa C , Fosforilación , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Mioblastos Esqueléticos/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 321(2): H382-H389, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34142888

RESUMEN

Pulmonary hypertension (PH) is associated with structural remodeling of pulmonary arteries (PAs) because of excessive proliferation of fibroblasts, endothelial cells, and smooth muscle cells (SMCs). The peptide hormone angiotensin II (ANG II) contributes to pulmonary vascular remodeling, in part, through its ability to trigger extracellular signal-regulated kinase (ERK1/2) activation. Here, we demonstrate that the ERK1/2 phosphatase, dual-specificity phosphatase 5 (DUSP5), functions as a negative regulator of ANG II-mediated SMC proliferation and PH. In contrast to wild-type controls, Dusp5 null mice infused with ANG II developed PH and right ventricular (RV) hypertrophy. PH in Dusp5 null mice was associated with thickening of the medial layer of small PAs, suggesting an in vivo role for DUSP5 as a negative regulator of ANG II-dependent SMC proliferation. Consistent with this, overexpression of DUSP5 blocked ANG II-mediated proliferation of cultured human pulmonary artery SMCs (hPASMCs) derived from patients with idiopathic PH or from failed donor controls. Collectively, the data support a role for DUSP5 as a feedback inhibitor of ANG II-mediated ERK signaling and PASMC proliferation and suggest that disruption of this circuit leads to adverse cardiopulmonary remodeling.NEW & NOTEWORTHY Dual-specificity phosphatases (DUSPs) serve critical roles in the regulation of mitogen-activated protein kinases, but their functions in the cardiovascular system remain poorly defined. Here, we provide evidence that DUSP5, which resides in the nucleus and specifically dephosphorylates extracellular signal-regulated kinase (ERK1/2), blocks pulmonary vascular smooth muscle cell proliferation. In response to angiotensin II infusion, mice lacking DUSP5 develop pulmonary hypertension and right ventricular cardiac hypertrophy. These findings illustrate DUSP5-mediated suppression of ERK signaling in the lungs as a protective mechanism.


Asunto(s)
Proliferación Celular/genética , Fosfatasas de Especificidad Dual/genética , Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/genética , Hipertrofia Ventricular Derecha/genética , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Remodelación Vascular/genética , Angiotensina II/farmacología , Animales , Estudios de Casos y Controles , Células Cultivadas , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/inducido químicamente , Hipertrofia Ventricular Derecha/fisiopatología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Vasoconstrictores/farmacología
6.
Molecules ; 26(5)2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33668293

RESUMEN

Plant-based foods, like fruits, vegetables, whole grains, legumes, nuts, seeds and other foodstuffs, have been deemed as heart healthy. The chemicals within these plant-based foods, i.e., phytochemicals, are credited with protecting the heart. However, the mechanistic actions of phytochemicals, which prevent clinical endpoints, such as pathological cardiac hypertrophy, are still being elucidated. We sought to characterize the overlapping and divergent mechanisms by which 18 selected phytochemicals prevent phenylephrine- and phorbol 12-myristate 13-acetate-mediated cardiomyocyte enlargement. Of the tested 18 compounds, six attenuated PE- and PMA-mediated enlargement of neonatal rat ventricular myocytes. Cell viability assays showed that apigenin, baicalein, berberine hydrochloride, emodin, luteolin and quercetin dihydrate did not reduce cell size through cytotoxicity. Four of the six phytochemicals, apigenin, baicalein, berberine hydrochloride and emodin, robustly inhibited stress-induced hypertrophy and were analyzed further against intracellular signaling and genome-wide changes in mRNA expression. The four phytochemicals differentially regulated mitogen-activated protein kinases and protein kinase D. RNA-sequencing further showed divergence in gene regulation, while pathway analysis demonstrated overlap in the regulation of inflammatory pathways. Combined, this study provided a comprehensive analysis of cardioprotective phytochemicals. These data highlight two defining observations: (1) that these compounds predominantly target divergent gene pathways within cardiac myocytes and (2) that regulation of overlapping signaling and gene pathways may be of particular importance for the anti-hypertrophic actions of these phytochemicals. Despite these new findings, future works investigating rodent models of heart failure are still needed to understand the roles for these compounds in the heart.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Cardiotónicos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Fitoquímicos/farmacología , Animales , Cardiomegalia/metabolismo , Cardiotónicos/química , Células Cultivadas , Miocitos Cardíacos/metabolismo , Fitoquímicos/química , Ratas , Ratas Sprague-Dawley
7.
Int J Mol Sci ; 21(11)2020 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-32503339

RESUMEN

Cardiovascular diseases (CVD) are the main cause of death worldwide and create a substantial financial burden. Emerging studies have begun to focus on epigenetic targets and re-establishing healthy gut microbes as therapeutic options for the treatment and prevention of CVD. Phytochemicals, commonly found in fruits and vegetables, have been shown to exert a protective effect against CVD, though their mechanisms of action remain incompletely understood. Of interest, phytochemicals such as curcumin, resveratrol and epigallocatechin gallate (EGCG) have been shown to regulate both histone acetylation and microbiome re-composition. The purpose of this review is to highlight the microbiome-epigenome axis as a therapeutic target for food bioactives in the prevention and/or treatment of CVD. Specifically, we will discuss studies that highlight how the three phytochemicals above alter histone acetylation leading to global changes in gene expression and CVD protection. Then, we will expand upon these phytochemicals to discuss the impact of phytochemical-microbiome-histone acetylation interaction in CVD.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Epigénesis Genética , Histonas/química , Microbiota , Acetilación , Animales , Enfermedades Cardiovasculares/microbiología , Catequina/análogos & derivados , Catequina/farmacología , Curcumina/farmacología , Histona Desacetilasas/metabolismo , Humanos , Fitoquímicos/farmacología , Procesamiento Proteico-Postraduccional , Resveratrol/farmacología
8.
Am J Physiol Endocrinol Metab ; 318(5): E765-E778, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32228320

RESUMEN

We report here that the neuronal (pro)renin receptor (PRR), a key component of the brain renin-angiotensin system (RAS), plays a critical role in the central regulation of high-fat-diet (HFD)-induced metabolic pathophysiology. The neuronal PRR is known to mediate formation of the majority of angiotensin (ANG) II, a key bioactive peptide of the RAS, in the central nervous system and to regulate blood pressure and cardiovascular function. However, little is known about neuronal PRR function in overnutrition-related metabolic physiology. Here, we show that PRR deletion in neurons reduces blood pressure, neurogenic pressor activity, and fasting blood glucose and improves glucose tolerance without affecting food intake or body weight following a 16-wk HFD. Mechanistically, we found that a HFD increases levels of the PRR ligand (pro)renin in the circulation and hypothalamus and of ANG II in the hypothalamus, indicating activation of the brain RAS. Importantly, PRR deletion in neurons reduced astrogliosis and activation of the astrocytic NF-κB p65 (RelA) in the arcuate nucleus and the ventromedial nucleus of the hypothalamus. Collectively, our findings indicate that the neuronal PRR plays essential roles in overnutrition-related metabolic pathophysiology.


Asunto(s)
Astrocitos/metabolismo , Glucemia/metabolismo , Presión Sanguínea/fisiología , Hipotálamo/metabolismo , Inflamación/metabolismo , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Peso Corporal/fisiología , Dieta Alta en Grasa , Ingestión de Alimentos/fisiología , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Renina/metabolismo , Receptor de Prorenina
9.
J Nutr Biochem ; 79: 108339, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32007664

RESUMEN

Pathological cardiac hypertrophy is a classical hallmark of heart failure. At the molecular level, inhibition of histone deacetylase (HDAC) enzymes attenuate pathological cardiac hypertrophy in vitro and in vivo. Emodin is an anthraquinone that has been implicated in cardiac protection. However, it is not known if the cardio-protective actions for emodin are mediated through HDAC-dependent regulation of gene expression. Therefore, we hypothesized that emodin would attenuate pathological cardiac hypertrophy via inhibition of HDACs, and that these actions would be reflected in an emodin-rich food like rhubarb. In this study, we demonstrate that emodin and Turkish rhubarb containing emodin inhibit HDAC activity in vitro, with fast-on, slow-off kinetics. Moreover, we show that emodin increased histone acetylation in cardiomyocytes concomitant to global changes in gene expression; gene expression changes were similar to the well-established pan-HDAC inhibitor trichostatin A (TSA). We additionally present evidence that emodin inhibited phenylephrine (PE) and phorbol myristate acetate (PMA)-induced hypertrophy in neonatal rat ventricular myocytes (NRVMs). Lastly, we demonstrate that the cardioprotective actions of emodin are translated to an angiotensin II (Ang) mouse model of cardiac hypertrophy and fibrosis and are linked to HDAC inhibition. These data suggest that emodin blocked pathological cardiac hypertrophy, in part, by inhibiting HDAC-dependent gene expression changes.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Emodina/farmacología , Histona Desacetilasas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Rheum/química , Acetilación , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Cardiomegalia/metabolismo , Cardiotónicos/farmacología , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Nat Prod Rep ; 37(5): 653-676, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31993614

RESUMEN

Covering: up to 2020Chronic, low-grade inflammation is linked to aging and has been termed "inflammaging". Inflammaging is considered a key contributor to the development of metabolic dysfunction and a broad spectrum of diseases or disorders including declines in brain and heart function. Genome-wide association studies (GWAS) coupled with epigenome-wide association studies (EWAS) have shown the importance of diet in the development of chronic and age-related diseases. Moreover, dietary interventions e.g. caloric restriction can attenuate inflammation to delay and/or prevent these diseases. Common themes in these studies entail the use of phytochemicals (plant-derived compounds) or the production of short chain fatty acids (SCFAs) as epigenetic modifiers of DNA and histone proteins. Epigenetic modifications are dynamically regulated and as such, serve as potential therapeutic targets for the treatment or prevention of age-related disease. In this review, we will focus on the role for natural products that include phytochemicals and short chain fatty acids (SCFAs) as regulators of these epigenetic adaptations. Specifically, we discuss regulators of methylation, acetylation and acylation, in the protection from chronic inflammation driven metabolic dysfunction and deterioration of neurocognitive and cardiac function.


Asunto(s)
Envejecimiento/genética , Productos Biológicos/farmacología , Inflamación/tratamiento farmacológico , Enfermedades Neurodegenerativas/prevención & control , Fitoquímicos/farmacología , Acetilación , Envejecimiento/efectos de los fármacos , Productos Biológicos/química , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Dieta , Epigénesis Genética , Ácidos Grasos Volátiles/farmacología , Humanos , Inflamación/etiología , Inflamación/genética , Enfermedades Neurodegenerativas/etiología
11.
Bioinformatics ; 36(6): 1663-1667, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31688895

RESUMEN

MOTIVATION: Our previous study has shown that ERBB2 is overexpressed in the organoid model of MCF10A when the stiffness of the microenvironment is increased to that of high mammographic density (MD). We now aim to identify key transcription factors (TFs) and functional enhancers that regulate processes associated with increased stiffness of the microenvironment in the organoid models of premalignant human mammary cell lines. RESULTS: 3D colony organizations and the cis-regulatory networks of two human mammary epithelial cell lines (184A1 and MCF10A) are investigated as a function of the increased stiffness of the microenvironment within the range of MD. The 3D colonies are imaged using confocal microscopy, and the morphometries of colony organizations and heterogeneity are quantified as a function of the stiffness of the microenvironment using BioSig3D. In a surrogate assay, colony organizations are profiled by transcriptomics. Transcriptome data are enriched by correlative analysis with the computed morphometric indices. Next, a subset of enriched data are processed against publicly available ChIP-Seq data using Model-based Analysis of Regulation of Gene Expression to predict regulatory transcription factors. This integrative analysis of morphometric and transcriptomic data predicted YY1 as one of the cis-regulators in both cell lines as a result of the increased stiffness of the microenvironment. Subsequent experiments validated that YY1 is expressed at protein and mRNA levels for MCF10A and 184A1, respectively. Also, there is a causal relationship between activation of YY1 and ERBB2 when YY1 is overexpressed at the protein level in MCF10A. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Densidad de la Mama , Organoides , Factor de Transcripción YY1 , Línea Celular , Biología Computacional , Humanos , Factores de Transcripción
12.
Future Med Chem ; 11(11): 1345-1356, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31161804

RESUMEN

Eight million US adults are projected to suffer from heart failure (HF) by 2030. Of concern, 5-year mortality rates following HF diagnosis approximate 40%. Small molecule histone deacetylase (HDAC) inhibitors have demonstrated efficacy for the treatment and reversal of HF. Historically, HDACs were studied as regulators of nucleosomal histones, in which lysine deacetylation on histone tails changed DNA-histone protein electrostatic interactions, leading to chromatin condensation and changes in gene expression. However, recent proteomics studies have demonstrated that approximately 4500 proteins can be acetylated in various tissues; the function of most of these remains unknown. This Review will focus on the nonepigenetic role for lysine acetylation in the heart, with a focus on nonepigenetic actions for HDAC inhibitors on cardiac function.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Acetilación/efectos de los fármacos , Animales , Corazón/efectos de los fármacos , Insuficiencia Cardíaca/metabolismo , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Lisina/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
13.
J Cell Physiol ; 234(2): 1088-1098, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30203485

RESUMEN

Bovine mammary epithelial cells (MAC-Ts) are a common cell line for the study of mammary epithelial inflammation; these cells are used to mechanistically elucidate molecular underpinnings that contribute to bovine mastitis. Bovine mastitis is the most prevalent form of disease in dairy cattle that culminates in annual losses of two billion dollars for the US dairy industry. Thus, there is an urgent need for improved therapeutic strategies. Histone deacetylase (HDAC) inhibitors are efficacious in rodent models of inflammation, yet their role in bovine mammary cells remain unclear. HDACs have traditionally been studied in the regulation of nucleosomal DNA, in which deacetylation of histones impact chromatin accessibility and gene expression. Using MAC-T cells stimulated with tumor necrosis factor α (TNF-α) as a model for mammary cell inflammation, we report that inhibition of HDACs1 and 2 (HDAC1/2) attenuated TNF-α-mediated inflammatory gene expression. Of note, we report that HDAC1/2-mediated inflammatory gene expression was partly regulated by c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) phosphorylation. Here, we report that HDAC1/2 inhibition attenuated JNK and ERK activation and thus inflammatory gene expression. These data suggest that HDACs1 and 2 regulate inflammatory gene expression via canonical (i.e., gene expression) and noncanonical (e.g., signaling dependent) mechanisms. Whereas, further studies using primary cell lines and animal models are needed. Our combined data suggest that HDAC1/2-specific inhibitors may prove efficacious for the treatment of bovine mastitis.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Glándulas Mamarias Animales/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antiinflamatorios/uso terapéutico , Bovinos , Línea Celular , Células Epiteliales/enzimología , Femenino , Regulación de la Expresión Génica , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 2/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Glándulas Mamarias Animales/enzimología , Mastitis Bovina/tratamiento farmacológico , Mastitis Bovina/enzimología , Fosforilación , Transducción de Señal
14.
Cell Signal ; 53: 234-245, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30347224

RESUMEN

Obesity is a strong predictor of heart disease, insulin resistance, and type II diabetes. Chronic, low-grade inflammation links obesity and insulin resistance through mitogen-activated protein kinase (MAPK) signaling pathways. Upstream kinases activate MAPK signaling, while MAPK-specific dual-specificity phosphatases (DUSPs) act as key modulators and controllers of MAPK deactivation (i.e. dephosphorylation). Using tumor necrosis factor α (TNFα) in 3 T3-L1 adipocytes as a model of inflammation, we report that TNFα-mediated induction of Dusp1, Dusp8 and Dusp16 modulated the transient regulation of MAPK (i.e., ERK, JNK, and p38) phosphorylation and subsequent inflammatory gene expression. All three MAPKs examined were phosphorylated in preadipocytes and adipocytes in response to TNFα, where signaling magnitude and duration were phenotype-specific. Moreover, TNFα increased mRNA abundance of DUSPs in preadipocytes and adipocytes in a phenotype-specific manner, concomitant with dephosphorylation of MAPKs. RNA interference (RNAi)-mediated knockdown of Dusp1, Dusp8 and Dusp16 increased signaling magnitude and duration of ERK, JNK, and p38 that subsequently resulted in significant increases in MAPK-dependent inflammatory gene expression of MCP-1, IL-6, and Cox-2 in response to TNFα. This study highlights important roles for DUSPs as integral components of MAPK signaling and adipocyte inflammatory gene expression.


Asunto(s)
Adipocitos/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , Células 3T3-L1 , Animales , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Anim Sci ; 96(12): 5244-5252, 2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30252114

RESUMEN

Histone deacetylase (HDAC) inhibition attenuates inflammation in rodents and short-chain fatty acids (SCFAs) are effective HDAC inhibitors. Therefore, the objective of this study was to evaluate the role of the SCFAs sodium propionate (SP) and sodium butyrate (SB) as HDAC-dependent regulators of inflammatory gene expression in bovine mammary epithelial cells (MAC-Ts). We postulated that SP and SB would decrease inflammation in MAC-Ts by inhibiting HDAC activity and increasing histone H3 acetylation and consequently decreasing inflammatory gene expression. For this study, MAC-Ts stimulated with lipopolysaccharide (LPS) were used as a model for bovine mammary epithelial cell inflammation. MAC-Ts were cultured in a basal medium. Cell lysates were incubated with SP or SB (0 to 5 mM) for 2 h prior to HDAC substrates incubation for an additional 2 h and HDACs activity was determined. Next, cells were pretreated with SP or SB (0 to 3.0 mM) for 2 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and assessed for histone H3 acetylation. Then, cells were pretreated with SP or SB (1 mM) for 24 h prior to LPS (1 µg/mL) stimulation for an additional 2 h and RNA was isolated for inflammatory gene expression evaluation by PCR array and gene validation was performed using quantitative real-time PCR. One-way ANOVA followed by Tukey post hoc analysis was conducted and statistical significance set at P < 0.05. SP and SB concentration-dependently and selectively inhibited class I HDAC activity, which differed between SCFAs, where SB inhibited (P < 0.05) HDACs 2, 3, and 8, while SP inhibited (P < 0.05) HDACs 2 and 8. Histone H3 acetylation was concentration-dependently increased by SCFAs and likewise the differential regulation of HDAC activity, SCFAs effected differently histone H3 acetylation, where SB increased (P < 0.05) H3K9/14, H3K18 and H3K27 acetylation, while SP increased (P < 0.05) H3K9/14 and H3K18 acetylation. However, SCFAs did not decrease (P > 0.05) overall inflammatory gene expression. Under our experimental conditions, findings suggest that in MAC-Ts, SCFAs regulate epigenetic markers on nucleosomal DNA in addition to regulation of inflammatory gene events independent of HDAC activity. Nevertheless, examination of SCFAs and/or HDACs inhibitors in bovine mammary gland is worth being further investigated to delineate the potential impact of HDAC inhibition and histones hyperacetylation on mammary gland tissue inflammation.


Asunto(s)
Ácido Butírico/farmacología , Bovinos/metabolismo , Ácidos Grasos Volátiles/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Mastitis Bovina/tratamiento farmacológico , Propionatos/farmacología , Acetilación/efectos de los fármacos , Animales , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Lipopolisacáridos/efectos adversos , Glándulas Mamarias Animales/metabolismo , Mastitis Bovina/inducido químicamente
16.
Nutrients ; 10(8)2018 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-30126190

RESUMEN

Approximately 5.7 million U.S. adults have been diagnosed with heart failure (HF). More concerning is that one in nine U.S. deaths included HF as a contributing cause. Current HF drugs (e.g., ß-blockers, ACEi) target intracellular signaling cascades downstream of cell surface receptors to prevent cardiac pump dysfunction. However, these drugs fail to target other redundant intracellular signaling pathways and, therefore, limit drug efficacy. As such, it has been postulated that compounds designed to target shared downstream mediators of these signaling pathways would be more efficacious for the treatment of HF. Histone deacetylation has been linked as a key pathogenetic element for the development of HF. Lysine residues undergo diverse and reversible post-translational modifications that include acetylation and have historically been studied as epigenetic modifiers of histone tails within chromatin that provide an important mechanism for regulating gene expression. Of recent, bioactive compounds within our diet have been linked to the regulation of gene expression, in part, through regulation of the epi-genome. It has been reported that food bioactives regulate histone acetylation via direct regulation of writer (histone acetyl transferases, HATs) and eraser (histone deacetylases, HDACs) proteins. Therefore, bioactive food compounds offer unique therapeutic strategies as epigenetic modifiers of heart failure. This review will highlight food bio-actives as modifiers of histone deacetylase activity in the heart.


Asunto(s)
Epigénesis Genética , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/genética , Inhibidores de Histona Desacetilasas/farmacología , Fitoquímicos/farmacología , Acetilación , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Regulación de la Expresión Génica , Interacción Gen-Ambiente , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Transducción de Señal
17.
Biosci Rep ; 38(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30061171

RESUMEN

Lysine residues undergo diverse and reversible post-translational modifications (PTMs). Lysine acetylation has traditionally been studied in the epigenetic regulation of nucleosomal histones that provides an important mechanism for regulating gene expression. Histone acetylation plays a key role in cardiac remodeling and function. However, recent studies have shown that thousands of proteins can be acetylated at multiple acetylation sites, suggesting the acetylome rivals the kinome as a PTM. Based on this, we examined the impact of obesity on protein lysine acetylation in the left ventricle (LV) of male c57BL/6J mice. We reported that obesity significantly increased heart enlargement and fibrosis. Moreover, immunoblot analysis demonstrated that lysine acetylation was markedly altered with obesity and that this phenomenon was cardiac tissue specific. Mass spectral analysis identified 2515 proteins, of which 65 were significantly impacted by obesity. Ingenuity Pathway Analysis® (IPA) further demonstrated that these proteins were involved in metabolic dysfunction and cardiac remodeling. In addition to total protein, 189 proteins were acetylated, 14 of which were significantly impacted by obesity. IPA identified the Cardiovascular Disease Pathway as significantly regulated by obesity. This network included aconitate hydratase 2 (ACO2), and dihydrolipoyl dehydrogenase (DLD), in which acetylation was significantly increased by obesity. These proteins are known to regulate cardiac function yet, the impact for ACO2 and DLD acetylation remains unclear. Combined, these findings suggest a critical role for cardiac acetylation in obesity-mediated remodeling; this has the potential to elucidate novel targets that regulate cardiac pathology.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Obesidad/metabolismo , Proteínas/metabolismo , Espectrometría de Masas en Tándem/métodos , Animales , Dieta Alta en Grasa/efectos adversos , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/fisiopatología , Procesamiento Proteico-Postraduccional , Proteínas/genética , Proteoma/análisis , Proteoma/metabolismo , Remodelación Ventricular
18.
JCI Insight ; 3(15)2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30089714

RESUMEN

Little is known about the biological function of histone deacetylase 11 (HDAC11), which is the lone class IV HDAC. Here, we demonstrate that deletion of HDAC11 in mice stimulates brown adipose tissue (BAT) formation and beiging of white adipose tissue (WAT). Consequently, HDAC11-deficient mice exhibit enhanced thermogenic potential and, in response to high-fat feeding, attenuated obesity, improved insulin sensitivity, and reduced hepatic steatosis. Ex vivo and cell-based assays revealed that HDAC11 catalytic activity suppresses the BAT transcriptional program, in both the basal state and in response to ß-adrenergic receptor signaling, through a mechanism that is dependent on physical association with BRD2, a bromodomain and extraterminal (BET) acetyl-histone-binding protein. These findings define an epigenetic pathway for the regulation of energy homeostasis and suggest the potential for HDAC11-selective inhibitors for the treatment of obesity and diabetes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Hígado Graso/patología , Histona Desacetilasas/metabolismo , Obesidad/patología , Termogénesis/genética , Factores de Transcripción/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Epigénesis Genética/fisiología , Hígado Graso/genética , Femenino , Regulación de la Expresión Génica/fisiología , Histona Desacetilasas/genética , Humanos , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Obesidad/genética
19.
Int J Mol Sci ; 20(1)2018 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-30597863

RESUMEN

Approximately five million United States (U.S.) adults are diagnosed with heart failure (HF), with eight million U.S. adults projected to suffer from HF by 2030. With five-year mortality rates following HF diagnosis approximating 50%, novel therapeutic treatments are needed for HF patients. Pre-clinical animal models of HF have highlighted histone deacetylase (HDAC) inhibitors as efficacious therapeutics that can stop and potentially reverse cardiac remodeling and dysfunction linked with HF development. HDACs remove acetyl groups from nucleosomal histones, altering DNA-histone protein electrostatic interactions in the regulation of gene expression. However, HDACs also remove acetyl groups from non-histone proteins in various tissues. Changes in histone and non-histone protein acetylation plays a key role in protein structure and function that can alter other post translational modifications (PTMs), including protein phosphorylation. Protein phosphorylation is a well described PTM that is important for cardiac signal transduction, protein activity and gene expression, yet the functional role for acetylation-phosphorylation cross-talk in the myocardium remains less clear. This review will focus on the regulation and function for acetylation-phosphorylation cross-talk in the heart, with a focus on the role for HDACs and HDAC inhibitors as regulators of acetyl-phosphorylation cross-talk in the control of cardiac function.


Asunto(s)
Histona Desacetilasas/metabolismo , Miocardio/metabolismo , Acetilación , Animales , Biomarcadores , Corazón/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional
20.
Curr Pharm Des ; 24(2): 158-170, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29189132

RESUMEN

BACKGROUND: Cardiovascular disease (CVD) is currently the leading cause of death globally. The metabolic syndrome (MetS), a clustering of risk factors including hypertension, hyperglycemia, elevated low-density lipoprotein (LDL) cholesterol, reduced high-density lipoprotein (HDL) cholesterol and increased visceral adiposity, is a significant risk factor for the development of CVD. Non-alcoholic fatty liver disease (NAFLD), often referred to as the hepatic manifestation of MetS, is a constellation of progressive liver disorders closely linked to obesity, diabetes, and insulin resistance. NAFLD initially presents as relatively benign, non-progressive hepatic steatosis, but it may, in certain individuals, progress to nonalcoholic steatohepatitis, fibrosis, cirrhosis, or hepatocellular carcinoma. Currently, there are no validated treatments for NAFLD. Polyphenols are important bioactive dietary compounds and may represent a natural complementary and integrative therapy for the treatment of CVDassociated risk factors, including elevated serum cholesterol and triglyceride levels, as well as NAFLD. Understanding their molecular mechanisms of action is important in the design of future human intervention studies. METHODS: Several studies utilizing in vitro and in vivo models have helped to identify underlying molecular mechanisms of action of polyphenols. RESULTS: This review will highlight recent advances regarding the molecular actions of dietary procyanidins, with a special focus on those originating from procyanidin-rich grape seed extracts, with a focus on the signaling pathways utilized to exert beneficial metabolic effects. CONCLUSION: Modulation of nuclear receptor activity and histone deacetylase inhibition has been identified as underlying mechanisms contributing to procyanidin-mediated amelioration of dyslipidemia and steatosis.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Polifenoles/metabolismo , Proantocianidinas/metabolismo , Transducción de Señal , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Polifenoles/administración & dosificación , Polifenoles/farmacología , Proantocianidinas/administración & dosificación , Proantocianidinas/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...