Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 23(11): 101728, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33241196

RESUMEN

Particulate matter ≤2.5µm (PM2.5) air pollution is a leading environmental risk factor contributing disproportionately to the global burden of non-communicable disease. We compared impact of chronic exposure to PM2.5 alone, or with light at night exposure (LL) on metabolism. PM2.5 induced peripheral insulin resistance, circadian rhythm (CR) dysfunction, and metabolic and brown adipose tissue (BAT) dysfunction, akin to LL (with no additive interaction between PM2.5 and LL). Transcriptomic analysis of liver and BAT revealed widespread but unique alterations in CR genes, with evidence for differentially accessible promoters and enhancers of CR genes in response to PM2.5 by ATAC-seq. The histone deacetylases 2, 3, and 4 were downregulated with PM2.5 exposure, with increased promoter occupancy by the histone acetyltransferase p300 as evidenced by ChIP-seq. These findings suggest a previously unrecognized role of PM2.5 in promoting CR disruption and metabolic dysfunction through epigenetic regulation of circadian targets.

2.
J Clin Invest ; 130(11): 6034-6040, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32780721

RESUMEN

Air pollution involving particulate matter smaller than 2.5 µm in size (PM2.5) is the world's leading environmental risk factor contributing to mortality through cardiometabolic pathways. In this study, we modeled early life exposure using chow-fed C57BL/6J male mice that were exposed to real-world inhaled, concentrated PM2.5 (~10 times ambient levels/~60-120 µg/m3) or filtered air over a 14-week period. We investigated the effects of PM2.5 on phenotype, the transcriptome, and chromatin accessibility and compared these with the effects of a prototypical high-fat diet (HFD) as well as cessation of exposure on phenotype reversibility. Exposure to PM2.5 impaired glucose and insulin tolerance and reduced energy expenditure and 18FDG-PET uptake in brown adipose tissue. Multiple differentially expressed gene clusters in pathways involving metabolism and circadian rhythm were noted in insulin-responsive tissues. Although the magnitude of transcriptional change detected with PM2.5 exposure was lower than that observed with a HFD, the degree of alteration in chromatin accessibility after PM2.5 exposure was significant. The novel chromatin remodeler SMARCA5 (SWI/SNF complex) was regulated in response to PM2.5 exposure, the cessation of which was associated with a reversal of insulin resistance and restoration of chromatin accessibility and nucleosome positioning near transcription start sites, as well as a reversal of exposure-induced changes in the transcriptome, including SMARCA5. These changes indicate pliable epigenetic control mechanisms following cessation of exposure.


Asunto(s)
Tejido Adiposo Pardo , Contaminantes Atmosféricos/toxicidad , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/efectos de los fármacos , Exposición a Riesgos Ambientales/efectos adversos , Resistencia a la Insulina , Adenosina Trifosfatasas/metabolismo , Tejido Adiposo Pardo/diagnóstico por imagen , Tejido Adiposo Pardo/metabolismo , Animales , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Proteínas Cromosómicas no Histona/metabolismo , Fluorodesoxiglucosa F18/farmacología , Ratones , Tomografía de Emisión de Positrones , Transcriptoma/efectos de los fármacos
3.
Arterioscler Thromb Vasc Biol ; 40(4): 914-928, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32078363

RESUMEN

OBJECTIVE: Systemic low-grade inflammation associated with obesity and metabolic syndrome is a strong risk factor for the development of diabetes mellitus and associated cardiovascular complications. This inflammatory state is caused by release of proinflammatory cytokines by macrophages, especially in adipose tissue. Long noncoding RNAs regulate macrophage activation and inflammatory gene networks, but their role in macrophage dysfunction during diet-induced obesity has been largely unexplored. Approach and Results: We sequenced total RNA from peritoneal macrophages isolated from mice fed either high-fat diet or standard diet and performed de novo transcriptome assembly to identify novel differentially expressed mRNAs and long noncoding RNAs. A top candidate long noncoding RNA, macrophage inflammation-suppressing transcript (Mist), was downregulated in both peritoneal macrophages and adipose tissue macrophages from high-fat diet-fed mice. GapmeR-mediated Mist knockdown in vitro and in vivo upregulated expression of genes associated with immune response and inflammation and increased modified LDL (low-density lipoprotein) uptake in macrophages. Conversely, Mist overexpression decreased basal and LPS (lipopolysaccharide)-induced expression of inflammatory response genes and decreased modified LDL uptake. RNA-pull down coupled with mass spectrometry showed that Mist interacts with PARP1 (poly [ADP]-ribose polymerase-1). Disruption of this RNA-protein interaction increased PARP1 recruitment and chromatin PARylation at promoters of inflammatory genes, resulting in increased gene expression. Furthermore, human orthologous MIST was also downregulated by proinflammatory stimuli, and its expression in human adipose tissue macrophages inversely correlated with obesity and insulin resistance. CONCLUSIONS: Mist is a novel protective long noncoding RNA, and its loss during obesity contributes to metabolic dysfunction and proinflammatory phenotype of macrophages via epigenetic mechanisms.


Asunto(s)
Inflamación/fisiopatología , Activación de Macrófagos/genética , Obesidad/genética , Obesidad/fisiopatología , ARN Largo no Codificante/fisiología , Tejido Adiposo/metabolismo , Animales , Línea Celular , LDL-Colesterol/metabolismo , Cromatina/genética , Citocinas/fisiología , Regulación hacia Abajo , Humanos , Metabolismo de los Lípidos/genética , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/fisiopatología , Ratones Endogámicos C57BL , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli ADP Ribosilación , Regulación hacia Arriba
5.
Mol Cell Biol ; 38(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29203644

RESUMEN

Macrophages are strategically distributed in mammalian tissues and play an essential role in priming the immune response. However, macrophages need to constantly strike a balance between activation and inhibition states to avoid a futile inflammatory reaction. Here, we identify the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as a potent repressor of macrophage proinflammatory activation. Gain- and loss-of-function studies revealed that CITED2 is required for optimal peroxisome proliferator-activated receptor gamma (PPARγ) activation and attendant select anti-inflammatory gene expression in macrophages. More importantly, deficiency of CITED2 resulted in significant attenuation of rosiglitazone-induced PPARγ activity, PPARγ recruitment to target gene promoters, and anti-inflammatory target gene expression in macrophages. Interestingly, deficiency of Cited2 strikingly heightened proinflammatory gene expression through stabilization of hypoxia-inducible factor 1 alpha (HIF1α) protein in macrophages. Further, overexpression of Egln3 or inhibition of HIF1α in Cited2-deficient macrophages completely reversed elevated proinflammatory cytokine/chemokine gene expression. Importantly, mice bearing a myeloid cell-specific deletion of Cited2 were highly susceptible to endotoxin-induced sepsis symptomatology and mortality. Collectively, our observations identify CITED2 as a novel negative regulator of macrophage proinflammatory activation that protects the host from inflammatory insults.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , PPAR gamma/metabolismo , Células RAW 264.7
6.
Am J Hypertens ; 31(2): 150-165, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29186297

RESUMEN

Noncoding RNAs (ncRNA) include a diverse range of functional RNA species-microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) being most studied in pathophysiology. Cardiovascular morbidity is associated with differential expression of myriad miRNAs; miR-21, miR-155, miR-126, miR-146a/b, miR-143/145, miR-223, and miR-221 are the top 9 most reported miRNAs in hypertension and atherosclerotic disease. A single miRNA may have hundreds of messenger RNA targets, which makes a full appreciation of the physiologic ramifications of such broad-ranging effects a challenge. miR-21 is the most prominent ncRNA associated with hypertension and atherosclerotic disease due to its role as a "mechano-miR", responding to arterial shear stresses. "Immuno-miRs", such as miR-155 and miR-223, affect cardiovascular disease (CVD) via regulation of hematopoietic cell differentiation, chemotaxis, and activation in response to many pro-atherogenic stimuli. "Myo-miRs", such as miR-1 and miR-133, affect cardiac muscle plasticity and remodeling in response to mechanical overload. This in-depth review analyzes observational and experimental reports of ncRNAs in CVD, including future applications of ncRNA-based strategies in diagnosis, prediction (e.g., survival and response to small molecule therapy), and biologic therapy.


Asunto(s)
Enfermedades Cardiovasculares , MicroARNs/clasificación , ARN no Traducido , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/terapia , Humanos , Terapia Molecular Dirigida
7.
Am J Nephrol ; 46(4): 298-314, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29017166

RESUMEN

BACKGROUND: The remarkable success of clinical trials in mineralocorticoid receptor (MR) inhibition in heart failure has driven research on the physiological and pathological role(s) of nonepithelial MR expression. MR is widely expressed in the cardiovascular system and is a major determinant of endothelial function, smooth muscle tone, vascular remodeling, fibrosis, and blood pressure. An important new dimension is the appreciation of the role MR plays in immune cells and target organ damage in the heart, kidney and vasculature, and in the development of insulin resistance. SUMMARY: The mechanism for MR activation in tissue injury continues to evolve with the evidence to date suggesting that activation of MR results in a complex repertoire of effects involving both macrophages and T cells. MR is an important transcriptional regulator of macrophage phenotype and function. Another important feature of MR activation is that it can occur even with normal or low aldosterone levels in pathological conditions. Tissue-specific conditional models of MR expression in myeloid cells, endothelial cells, smooth muscle cells and cardiomyocytes have been very informative and have firmly demonstrated a critical role of MR as a key pathophysiologic variable in cardiac hypertrophy, transition to heart failure, adipose inflammation, and atherosclerosis. Finally, the central nervous system activation of MR in permeable regions of the blood-brain barrier may play a role in peripheral inflammation. Key Message: Ongoing clinical trials will help clarify the role of MR blockade in conditions, such as atherosclerosis and chronic kidney disease.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inflamación/patología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Receptores de Mineralocorticoides/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Aterosclerosis/patología , Barrera Hematoencefálica/metabolismo , Ensayos Clínicos como Asunto , Humanos , Inflamación/tratamiento farmacológico , Riñón/metabolismo , Macrófagos/metabolismo , Miocardio/metabolismo , Miocitos del Músculo Liso/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal , Linfocitos T/metabolismo
8.
PLoS One ; 11(11): e0165962, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27812198

RESUMEN

Obesity in humans and mice is typified by an activated macrophage phenotype in the visceral adipose tissue (VAT) leading to increased macrophage-mediated inflammation. microRNAs (miRNAs) play an important role in regulating inflammatory pathways in macrophages, and in this study we compared miRNA expression in the VAT of insulin resistant morbidly obese humans to a non-obese cohort with normal glucose tolerance. miR-223-3p was found to be significantly upregulated in the whole omental tissue RNA of 12 human subjects, as were 8 additional miRNAs. We then confirmed that miR-223 upregulation was specific to the stromal vascular cells of human VAT, and found that miR-223 levels were unchanged in adipocytes and circulating monocytes of the non-obese and obese. miR-223 ablation increased basal / unstimulated TLR4 and STAT3 expression and LPS-stimulated TLR4, STAT3, and NOS2 expression in primary macrophages. Conversely, miR-223 mimics decreased TLR4 expression in primary macrophage, at the same time it negatively regulated FBXW7 expression, a well described suppressor of Toll-like receptor 4 (TLR4) signaling. We concluded that the abundance of miR-223 in macrophages significantly modulates macrophage phenotype / activation state and response to stimuli via effects on the TLR4/FBXW7 axis.


Asunto(s)
Grasa Intraabdominal/metabolismo , Macrófagos/inmunología , MicroARNs/genética , Obesidad/genética , Obesidad/inmunología , Regulación hacia Arriba , Adulto , Animales , Proteínas de Ciclo Celular/metabolismo , Estudios de Cohortes , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina , Activación de Macrófagos , Masculino , Ratones , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo II/metabolismo , Obesidad/patología , Fenotipo , Receptor Toll-Like 4/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
9.
Adv Drug Deliv Rev ; 99(Pt A): 2-11, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826436

RESUMEN

Clearance of apoptotic debris is a vital role of the innate immune system. Drawing upon principles of apoptotic clearance, convenient delivery vehicles including intrinsic anti-inflammatory characteristics and specificity to immune cells can be engineered to aid in drug delivery. In this article, we examine the use of phosphatidylserine (PtdSer), the well-known "eat-me" signal, in nanoparticle-based therapeutics making them highly desirable "meals" for phagocytic immune cells. Use of PtdSer facilitates engulfment of nanoparticles allowing for imaging and therapy in various pathologies and may result in immunomodulation. Furthermore, we discuss the targeting of the macrophages and other cells at sites of inflammation in disease. A thorough understanding of the immunobiology of "eat-me" signals is requisite for the successful application of "eat-me"-bearing materials in biomedical applications.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Fosfatidilserinas/administración & dosificación , Animales , Diagnóstico por Imagen , Quimioterapia , Humanos , Inmunidad Innata , Preparaciones Farmacéuticas/administración & dosificación
10.
J Control Release ; 217: 243-55, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26386437

RESUMEN

Macrophages are innate immune cells with great phenotypic plasticity, which allows them to regulate an array of physiological processes such as host defense, tissue repair, and lipid/lipoprotein metabolism. In this proof-of-principle study, we report that macrophages of the M1 inflammatory phenotype can be selectively targeted by model hybrid lipid-latex (LiLa) nanoparticles bearing phagocytic signals. We demonstrate a simple and robust route to fabricate nanoparticles and then show their efficacy through imaging and drug delivery in inflammatory disease models of atherosclerosis and obesity. Self-assembled LiLa nanoparticles can be modified with a variety of hydrophobic entities such as drug cargos, signaling lipids, and imaging reporters resulting in sub-100nm nanoparticles with low polydispersities. The optimized theranostic LiLa formulation with gadolinium, fluorescein and "eat-me" phagocytic signals (Gd-FITC-LiLa) a) demonstrates high relaxivity that improves magnetic resonance imaging (MRI) sensitivity, b) encapsulates hydrophobic drugs at up to 60% by weight, and c) selectively targets inflammatory M1 macrophages concomitant with controlled release of the payload of anti-inflammatory drug. The mechanism and kinetics of the payload discharge appeared to be phospholipase A2 activity-dependent, as determined by means of intracellular Förster resonance energy transfer (FRET). In vivo, LiLa targets M1 macrophages in a mouse model of atherosclerosis, allowing noninvasive imaging of atherosclerotic plaque by MRI. In the context of obesity, LiLa particles were selectively deposited to M1 macrophages within inflamed adipose tissue, as demonstrated by single-photon intravital imaging in mice. Collectively, our results suggest that phagocytic signals can preferentially target inflammatory macrophages in experimental models of atherosclerosis and obesity, thus opening the possibility of future clinical applications that diagnose/treat these conditions. Tunable LiLa nanoparticles reported here can serve as a model theranostic platform with application in various types of imaging of the diseases such as cardiovascular disorders, obesity, and cancer where macrophages play a pathogenic role.


Asunto(s)
Antiinflamatorios/administración & dosificación , Macrófagos/efectos de los fármacos , Nanopartículas/administración & dosificación , Animales , Antiinflamatorios/química , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Línea Celular , Colesterol/análogos & derivados , Colesterol/química , Citocinas/genética , Fluoresceína-5-Isotiocianato/química , Gadolinio/química , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/química , Obesidad/inmunología , Paclitaxel/administración & dosificación , Paclitaxel/química , Fagocitosis , Fosfatidiletanolaminas/química , Fosfatidilserinas/química , Fosfolipasas A2/química , Polietilenglicoles/química , Poliestirenos/química , Rosiglitazona , Tamoxifeno/administración & dosificación , Tamoxifeno/química , Tiazolidinedionas/administración & dosificación , Tiazolidinedionas/química
11.
Diabetes ; 63(4): 1289-302, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24222350

RESUMEN

A key pathophysiologic role for activated T-cells in mediating adipose inflammation and insulin resistance (IR) has been recently postulated. However, mechanisms underlying their activation are poorly understood. In this study, we demonstrated a previously unrecognized homeostatic role for the costimulatory B7 molecules (CD80 and CD86) in preventing adipose inflammation. Instead of promoting inflammation, which was found in many other disease conditions, B7 costimulation reduced adipose inflammation by maintaining regulatory T-cell (Treg) numbers in adipose tissue. In both humans and mice, expression of CD80 and CD86 was negatively correlated with the degree of IR and adipose tissue macrophage infiltration. Decreased B7 expression in obesity appeared to directly impair Treg proliferation and function that lead to excessive proinflammatory macrophages and the development of IR. CD80/CD86 double knockout (B7 KO) mice had enhanced adipose macrophage inflammation and IR under both high-fat and normal diet conditions, accompanied by reduced Treg development and proliferation. Adoptive transfer of Tregs reversed IR and adipose inflammation in B7 KO mice. Our results suggest an essential role for B7 in maintaining Tregs and adipose homeostasis and may have important implications for therapies that target costimulation in type 2 diabetes.


Asunto(s)
Tejido Adiposo/patología , Antígeno B7-1/fisiología , Antígeno B7-2/fisiología , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/fisiología , Tejido Adiposo/inmunología , Traslado Adoptivo , Animales , Proliferación Celular , Homeostasis/fisiología , Humanos , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Macrófagos/inmunología , Ratones , Ratones Noqueados
12.
Obesity (Silver Spring) ; 22(5): 1264-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24124129

RESUMEN

OBJECTIVE: Chemokine (C-X-C motif) receptor 3 (CXCR3) is a chemokine receptor involved in the regulation of immune cell trafficking and activation. Increased CXCR3 expression in the visceral adipose of obese humans and mice was observed. A pathophysiologic role for CXCR3 in diet-induced obesity (DIO) was hypothesized. METHODS: Wild-type (WT) C57B/L6J and chemokine receptor 3 knockout (CXCR3(-/-) ) mice were fed a high-fat diet (HFD) for 20 weeks followed by assessment of glucose metabolism and visceral adipose tissue (VAT) inflammation. RESULTS: CXCR3(-/-) mice exhibited lower fasting glucose and improved glucose tolerance compared with WT-HFD mice, despite similar body mass. HFD-induced VAT innate and adaptive immune cell infiltration, including immature myeloid cells (CD11b(+) F4/80(lo) Ly6C(+) ), were markedly ameliorated in CXCR3(-/-) mice. In vitro IBIDI and in vivo migration assays demonstrated no CXCR3-mediated effect on macrophage or monocyte migration, respectively. CXCR3(-/-) macrophages, however, had a blunted response to interferon-γ, a TH 1 cytokine that induces macrophage activation. CONCLUSIONS: A previously unreported role for CXCR3 in the development of HFD-induced insulin resistance (IR) and VAT macrophage infiltration in mice was demonstrated. Our results support pharmaceutical targeting of the CXCR3 receptor as a potential treatment for obesity/IR.


Asunto(s)
Resistencia a la Insulina , Grasa Intraabdominal/fisiopatología , Obesidad/fisiopatología , Receptores CXCR3/metabolismo , Adulto , Animales , Glucemia/metabolismo , Índice de Masa Corporal , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta Alta en Grasa , Marcación de Gen , Humanos , Inflamación/genética , Inflamación/metabolismo , Insulina/sangre , Interferón gamma/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , Receptores CXCR3/genética , Transducción de Señal , Triglicéridos/sangre , Regulación hacia Arriba
13.
Part Fibre Toxicol ; 9: 40, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23126276

RESUMEN

BACKGROUND: It has been well recognized that toxicity of fine ambient air particulate matter (PM(2.5)) may depend on its chemical constituents, including components such as soluble metals that may theoretically exert distinctive effects. We have recently demonstrated an important effect of PM(2.5) on metabolic function. Since transition metals, such as nickel (Ni), represent an important component of exposure in certain environments, and may significantly influence the toxicity of inhalational exposure, we investigated the effects of Ni as a variable component of ambient PM(2.5) exposure. METHODS: Male ApoE knockout mice were exposed to filtered air (FA), fine-sized nickel sulfate particles alone (Ni) at 0.44 µg/m(3), concentrated ambient air PM(2.5) (CAPs) at a mean of 70 µg/m(3), or CAPs+Ni in Tuxedo, NY, 6 hours/day, 5 days/week, for 3 months. RESULTS: Exposure to Ni, irrespective of co-exposure to CAPs, resulted in body weight gain, while exposure to CAPs+Ni significantly enhanced fasting glucose and worsened insulin resistance measures (HOMA-IR), when compared with exposure to CAPs alone. CAPs+Ni exposure induced a significant decrease in phosphorylation of AMP-activated protein kinase (AMPK) α. Exposure to Ni or CAPs+Ni significantly induced microcirculatory dysfunction and increased monocytic cell infiltration into lung and adipose, and decreased uncoupling protein 1 expression at gene and protein levels and several brown adipocyte-specific genes in adipose tissue. CONCLUSIONS: Ni exposure has effects on metabolic and inflammatory parameters that are comparable to that of CAPs. Additionally, Ni synergistically exacerbates CAPs-induced adverse effects on some of, but not all of, these parameters, that may be mediated via the AMPK signaling pathway. These findings have important implications for inhaled transition metal toxicity that may exert synergistic effects with other PM(2.5) components.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Exposición por Inhalación/efectos adversos , Resistencia a la Insulina , Mitocondrias/efectos de los fármacos , Níquel/toxicidad , Material Particulado/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/inmunología , Adipocitos/metabolismo , Animales , Apolipoproteínas E/genética , Glucemia/análisis , Citocinas/sangre , Sinergismo Farmacológico , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina/inmunología , Canales Iónicos/genética , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Tamaño Mitocondrial/efectos de los fármacos , Tamaño de la Partícula , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Desacopladora 1
14.
Arterioscler Thromb Vasc Biol ; 32(4): 962-70, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22308043

RESUMEN

OBJECTIVE: Myeloid-related protein (Mrp) 8/14 complex (is a highly expressed extracellularly secreted protein, implicated in atherosclerosis. In this study, we evaluated the feasibility of targeting Mrp in vivo through synthetic immuno-nanoprobes. METHODS AND RESULTS: Anti-Mrp-14 and nonspecific IgG-conjugated gadolinium nanoprobes (aMrp-) were synthesized and characterized. Pharmacokinetics and vascular targeting via MRI of the formulations were assessed in vivo in high fat-fed apolipoprotein E deficient (ApoE(-/-)), ApoE(-/-)/Mrp14(-/-) (double knockout) and chow-fed wild-type (C57BL/6) mice. Bone marrow-derived myeloid progenitor cells were isolated from both ApoE(-/-) and double knockout mice, differentiated to macrophages, and were treated with LPS, with or without Mrp8, Mrp14, or Mrp8/14; conditioned media was used for in vitro studies. Mrp-activated cells secreted significant amounts of proinflammatory cytokines, which was abolished by pretreatment with aMrp-NP. We show in vitro that aMrp-NP binds endothelial cells previously treated with conditioned media containing Mrp8/14. MRI following intravenous delivery of aMrp-NP revealed prolonged and substantial delineation of plaque in ApoE(-/-) but not double knockout or wild-type animals. Nonspecific IgG-conjugated gadolinium nanoprobe-injected animals in all groups did not show vessel wall enhancement. Flow-cytometric analysis of aortic digesta revealed that aMrp-NP present in Ly-6G(+), CD11b(+), CD11c(+), and CD31(+) cells in ApoE(-/-) but not in double knockout animals. CONCLUSIONS: Targeted imaging with aMrp-NP demonstrates enhancement of plaque with binding to inflammatory cells and reduction in inflammation. This strategy has promise as a theranostic approach for atherosclerosis.


Asunto(s)
Albúminas/farmacocinética , Antiinflamatorios/farmacocinética , Anticuerpos/metabolismo , Aterosclerosis/metabolismo , Calgranulina A/inmunología , Calgranulina B/inmunología , Medios de Contraste/farmacocinética , Gadolinio DTPA/farmacocinética , Inmunoconjugados/farmacocinética , Inflamación/metabolismo , Nanopartículas del Metal , Albúminas/química , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Anticuerpos/química , Anticuerpos/farmacología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/diagnóstico , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/inmunología , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Medios de Contraste/química , Medios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Estudios de Factibilidad , Citometría de Flujo , Gadolinio DTPA/química , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoconjugados/química , Inflamación/diagnóstico , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Progenitoras Mieloides/metabolismo , Distribución Tisular
15.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L399-409, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22160305

RESUMEN

The purpose of this study was to investigate the effects of chronically inhaled particulate matter <2.5 µm (PM(2.5)) on inflammatory cell populations in the lung and systemic circulation. A prominent component of air pollution exposure is a systemic inflammatory response that may exaggerate chronic diseases such as atherosclerosis and insulin resistance. T cell response was measured in wild-type C57B/L6, Foxp3-green fluorescent protein (GFP) "knockin," and chemokine receptor 3 knockout (CXCR3(-/-)) mice following 24-28 wk of PM(2.5) or filtered air. Chronic PM(2.5) exposure resulted in increased CXCR3-expressing CD4(+) and CD8(+) T cells in the lungs, spleen, and blood with elevation in CD11c(+) macrophages in the lung and oxidized derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine in wild-type mice. CXCR3 deficiency decreased T cells in the lung. GFP(+) regulatory T cells increased with PM(2.5) exposure in the spleen and blood of Foxp3-GFP mice but were present at very low levels in the lung irrespective of PM(2.5) exposure. Mixed lymphocyte cultures using primary, PM(2.5)-treated macrophages demonstrated enhanced T cell proliferation. Our experiments indicate that PM(2.5) potentiates a proinflammatory Th1 response involving increased homing of CXCR3(+) T effector cells to the lung and modulation of systemic T cell populations.


Asunto(s)
Contaminación del Aire , Activación de Linfocitos , Material Particulado/toxicidad , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Recuento de Células , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Expresión Génica , Inmunidad Innata , Interleucina-17/genética , Interleucina-17/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ganglios Linfáticos/patología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Material Particulado/inmunología , Fosfolípidos/metabolismo , Regiones Promotoras Genéticas , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Linfocitos T Reguladores
16.
Circulation ; 124(21): 2338-49, 2011 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22007077

RESUMEN

BACKGROUND: Dipeptidyl-peptidase 4 (DPP-4) inhibitors are increasingly used to accomplish glycemic targets in patients with type II diabetes mellitus. Because DPP-4 is expressed in inflammatory cells, we hypothesized that its inhibition will exert favorable effects in atherosclerosis. METHODS AND RESULTS: Male LDLR(-/-) mice (6 weeks) were fed a high-fat diet or normal chow diet for 4 weeks and then randomized to vehicle or alogliptin, a high-affinity DPP-4 inhibitor (40 mg · kg(-1) · d(-1)), for 12 weeks. Metabolic parameters, blood pressure, vascular function, atherosclerosis burden, and indexes of inflammation were obtained in target tissues, including the vasculature, adipose, and bone marrow, with assessment of global and cell-specific inflammatory pathways. In vitro and in vivo assays of DPP-4 inhibition (DPP-4i) on monocyte activation/migration were conducted in both human and murine cells and in a short-term ApoE(-/-) mouse model. DPP-4i improved markers of insulin resistance and reduced blood pressure. DPP-4i reduced visceral adipose tissue macrophage content (adipose tissue macrophages; CD11b(+), CD11c(+), Ly6C(hi)) concomitant with upregulation of CD163. DPP-4 was highly expressed in bone marrow-derived CD11b(+) cells, with DPP-4i downregulating proinflammatory genes in these cells. DPP-4i decreased aortic plaque with a striking reduction in plaque macrophages. DPP-4i prevented monocyte migration and actin polymerization in in vitro assays via Rac-dependent mechanisms and prevented in vivo migration of labeled monocytes to the aorta in response to exogenous tumor necrosis factor-α and DPP-4. CONCLUSION: DPP-4i exerts antiatherosclerotic effects and reduces inflammation via inhibition of monocyte activation/chemotaxis. These findings have important implications for the use of this class of drugs in atherosclerosis.


Asunto(s)
Aterosclerosis/patología , Aterosclerosis/prevención & control , Quimiotaxis/fisiología , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Inflamación/patología , Inflamación/prevención & control , Monocitos/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/fisiopatología , Presión Sanguínea/efectos de los fármacos , Movimiento Celular/fisiología , Quimiotaxis/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Modelos Animales de Enfermedad , Glucosa/metabolismo , Inflamación/fisiopatología , Resistencia a la Insulina/fisiología , Masculino , Metabolismo/efectos de los fármacos , Ratones , Ratones Noqueados , Piperidinas/farmacología , Piperidinas/uso terapéutico , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Tiempo , Uracilo/análogos & derivados , Uracilo/farmacología , Uracilo/uso terapéutico
17.
Circ Res ; 108(6): 716-26, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21273555

RESUMEN

RATIONALE: Chronic exposure to ambient air-borne particulate matter of < 2.5 µm (PM2.5) increases cardiovascular risk. The mechanisms by which inhaled ambient particles are sensed and how these effects are systemically transduced remain elusive. OBJECTIVE: To investigate the molecular mechanisms by which PM2.5 mediates inflammatory responses in a mouse model of chronic exposure. METHODS AND RESULTS: Here, we show that chronic exposure to ambient PM2.5 promotes Ly6C(high) inflammatory monocyte egress from bone-marrow and mediates their entry into tissue niches where they generate reactive oxygen species via NADPH oxidase. Toll-like receptor (TLR)4 and Nox2 (gp91(phox)) deficiency prevented monocyte NADPH oxidase activation in response to PM2.5 and was associated with restoration of systemic vascular dysfunction. TLR4 activation appeared to be a prerequisite for NAPDH oxidase activation as evidenced by reduced p47(phox) phosphorylation in TLR4 deficient animals. PM2.5 exposure markedly increased oxidized phospholipid derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine (oxPAPC) in bronchioalveolar lavage fluid. Correspondingly, exposure of bone marrow-derived macrophages to oxPAPC but not PAPC recapitulated effects of chronic PM2.5 exposure, whereas TLR4 deficiency attenuated this response. CONCLUSIONS: Taken together, our findings suggest that PM2.5 triggers an increase in oxidized phospholipids in lungs that then mediates a systemic cellular inflammatory response through TLR4/NADPH oxidase-dependent mechanisms.


Asunto(s)
NADPH Oxidasas/metabolismo , Material Particulado/efectos adversos , Receptor Toll-Like 4/metabolismo , Enfermedades Vasculares/inducido químicamente , Enfermedades Vasculares/etiología , Administración por Inhalación , Contaminantes Atmosféricos/efectos adversos , Animales , Exposición a Riesgos Ambientales , Activación Enzimática , Inflamación/etiología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Oxidación-Reducción , Tamaño de la Partícula , Material Particulado/administración & dosificación , Fosfolípidos/metabolismo , Factores de Tiempo
18.
Lipids ; 45(6): 479-89, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20509000

RESUMEN

Mice exposed to cold rely upon brown adipose tissue (BAT)-mediated nonshivering thermogenesis to generate body heat using dietary glucose and lipids from the liver and white adipose tissue. In this report, we investigate how cold exposure affects the PI3 K/Akt signaling cascade and the expression of genes involved in lipid metabolism and trafficking in BAT. Cold exposure at an early time point led to the activation of the PI3 K/Akt, insulin-like signaling cascade followed by a transient decrease in adipose triglyceride lipase (ATGL) gene and protein expression in BAT. To further investigate how cold exposure-induced signaling altered ATGL expression, cultured primary brown adipocytes were treated with the beta(3)-adrenergic receptor (beta(3)AR) agonist CL 316,243 (CL) resulting in activation of PI3 K/Akt, ERK 1/2, and p38 signaling pathways and significantly decreased ATGL protein levels. ATGL protein levels decreased significantly 30 min post CL treatment suggesting protein degradation. Inhibition of PKA signaling by H89 rescued ATGL levels. The effects of PKA signaling on ATGL were shown to be independent of relevant pathways downstream of PKA such as PI3 K/Akt, ERK 1/2, and p38. However, CL treatment in 3T3-L1 adipocytes did not decrease ATGL protein and mRNA expression, suggesting a distinct response in WAT to beta3-adrenergic agonism. Transitory effects, possibly attributed to acute Akt activation during the early recruitment phase, were noted as well as stable changes in gene expression which may be attributed to beta3-adrenergic signaling in BAT.


Asunto(s)
Adipocitos Marrones/enzimología , Regulación hacia Abajo , Lipasa/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Dioxoles , Lipasa/genética , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Adrenérgicos beta 3/genética
19.
Microvasc Res ; 78(3): 294-300, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19682464

RESUMEN

In this study, we describe the use of intravital microscopy in a transgenic mouse model expressing yellow fluorescent protein (YFP) under the control of a monocyte specific promoter c-fms (CD115) to track and quantify specific leukocyte subsets. Flow cytometry on peripheral and bone marrow leukocytes revealed that YFP was predominantly expressed by CD11a(+), CD11b(+), and CD14(+) monocytes. In the bone marrow, 67+/-4% of Ly6C(high) F4/80(+) cells were YFP(high) while 55+/-1% of Ly6C(low) F4/80(+) cells were YFP(low) supporting the use of c-fms(YFP) expression as a marker of monocyte lineage. 70+/-7% of CD11b(+) F4/80(+) Ly6C(+) ("triple positive") cells expressed YFP. To assess leukocyte-endothelial interactions in YFP(+) cells in c-fms(YFP+) mice, we evaluated leukocyte adhesion, rolling and local shear stress responses in the cremasteric endothelium 4 h following administration of TNFalpha. TNFalpha resulted in a five-fold increase in adhesion of YFP(+) cells to the endothelium and provided superior discriminative ability in assessing rolling and adhesion events when compared with bright field microscopy. Additionally, when compared with Rhodamine-6G labeled leukocytes or GFP(+) cells in mice transplanted with green fluorescent protein (GFP) positive bone marrow, the level of detail observed in the c-fms(YFP+) was greater, with both GFP(+) and YFP(+) cells demonstrating superior signal to noise compared to bright field microscopy. A weak positive linear correlation between wall shear stress and YFP(+) cell adhesion (r(2)=0.20, p<0.05) was seen in the cremasteric microcirculation. Taken together, these data demonstrate the use of c-fms(YFP+) mice in identifying distinct monocyte subsets and highlight the potential of this model for real-time monocyte-endothelial interactions using intravital microscopy.


Asunto(s)
Células de la Médula Ósea/citología , Células Endoteliales/citología , Endotelio Vascular/citología , Proteínas Luminiscentes/metabolismo , Subgrupos Linfocitarios/citología , Modelos Animales , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Adhesión Celular/fisiología , Comunicación Celular , Linaje de la Célula , Células Endoteliales/metabolismo , Endotelio Vascular/fisiología , Citometría de Flujo , Hematopoyesis/fisiología , Rodamiento de Leucocito/fisiología , Subgrupos Linfocitarios/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcirculación/fisiología , Microscopía por Video , Músculo Esquelético/irrigación sanguínea
20.
Circulation ; 119(4): 538-46, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19153269

RESUMEN

BACKGROUND: There is a strong link between urbanization and type 2 diabetes mellitus. Although a multitude of mechanisms have been proposed, there are no studies evaluating the impact of ambient air pollutants and the propensity to develop type 2 diabetes mellitus. We hypothesized that exposure to ambient fine particulate matter (<2.5 mum; PM(2.5)) exaggerates diet-induced insulin resistance, adipose inflammation, and visceral adiposity. METHODS AND RESULTS: Male C57BL/6 mice were fed high-fat chow for 10 weeks and randomly assigned to concentrated ambient PM(2.5) or filtered air (n=14 per group) for 24 weeks. PM(2.5)-exposed C57BL/6 mice exhibited marked whole-body insulin resistance, systemic inflammation, and an increase in visceral adiposity. PM(2.5) exposure induced signaling abnormalities characteristic of insulin resistance, including decreased Akt and endothelial nitric oxide synthase phosphorylation in the endothelium and increased protein kinase C expression. These abnormalilties were associated with abnormalities in vascular relaxation to insulin and acetylcholine. PM(2.5) increased adipose tissue macrophages (F4/80(+) cells) in visceral fat expressing higher levels of tumor necrosis factor-alpha/interleukin-6 and lower interleukin-10/N-acetyl-galactosamine specific lectin 1. To test the impact of PM(2.5) in eliciting direct monocyte infiltration into fat, we rendered FVBN mice expressing yellow fluorescent protein (YFP) under control of a monocyte-specific promoter (c-fms, c-fms(YFP)) diabetic over 10 weeks and then exposed these mice to PM(2.5) or saline intratracheally. PM(2.5) induced YFP cell accumulation in visceral fat and potentiated YFP cell adhesion in the microcirculation. CONCLUSIONS: PM(2.5) exposure exaggerates insulin resistance and visceral inflammation/adiposity. These findings provide a new link between air pollution and type 2 diabetes mellitus.


Asunto(s)
Contaminación del Aire/efectos adversos , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Inflamación/complicaciones , Obesidad/inmunología , Obesidad/metabolismo , Animales , Adhesión Celular/inmunología , Diabetes Mellitus Tipo 2/epidemiología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Exposición a Riesgos Ambientales , Inflamación/epidemiología , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Proteínas Luminiscentes/genética , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/epidemiología , Factores de Riesgo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...