Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 155(4): 793-806, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24209618

RESUMEN

The eukaryotic biological clock involves a negative transcription-translation feedback loop in which clock genes regulate their own transcription and that of output genes of metabolic significance. While around 10% of the liver transcriptome is rhythmic, only about a fifth is driven by de novo transcription, indicating mRNA processing is a major circadian component. Here, we report that inhibition of transmethylation reactions elongates the circadian period. RNA sequencing then reveals methylation inhibition causes widespread changes in the transcription of the RNA processing machinery, associated with m(6)A-RNA methylation. We identify m(6)A sites on many clock gene transcripts and show that specific inhibition of m(6)A methylation by silencing of the m(6)A methylase Mettl3 is sufficient to elicit circadian period elongation and RNA processing delay. Analysis of the circadian nucleocytoplasmic distribution of clock genes Per2 and Arntl then revealed an uncoupling between steady-state pre-mRNA and cytoplasmic mRNA rhythms when m(6)A methylation is inhibited.


Asunto(s)
Relojes Circadianos , Metiltransferasas/metabolismo , Procesamiento Postranscripcional del ARN , ARN/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Línea Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/genética , Proteínas Circadianas Period/metabolismo , Tubercidina/farmacología
2.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34426497

RESUMEN

Skeletal muscle atrophy is caused by various conditions, including aging, disuse related to a sedentary lifestyle and lack of physical activity, and cachexia. Our insufficient understanding of the molecular mechanism underlying muscle atrophy limits the targets for the development of effective pharmacologic treatments and preventions. Here, we identified Krüppel-like factor 5 (KLF5), a zinc-finger transcription factor, as a key mediator of the early muscle atrophy program. KLF5 was up-regulated in atrophying myotubes as an early response to dexamethasone or simulated microgravity in vitro. Skeletal muscle-selective deletion of Klf5 significantly attenuated muscle atrophy induced by mechanical unloading in mice. Transcriptome- and genome-wide chromatin accessibility analyses revealed that KLF5 regulates atrophy-related programs, including metabolic changes and E3-ubiquitin ligase-mediated proteolysis, in coordination with Foxo1. The synthetic retinoic acid receptor agonist Am80, a KLF5 inhibitor, suppressed both dexamethasone- and microgravity-induced muscle atrophy in vitro and oral Am80 ameliorated disuse- and dexamethasone-induced atrophy in mice. Moreover, in three independent sets of transcriptomic data from human skeletal muscle, KLF5 expression significantly increased with age and the presence of sarcopenia and correlated positively with the expression of the atrophy-related ubiquitin ligase genes FBXO32 and TRIM63 These findings demonstrate that KLF5 is a key transcriptional regulator mediating muscle atrophy and that pharmacological intervention with Am80 is a potentially preventive treatment.


Asunto(s)
Benzoatos/farmacología , Desarrollo de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/fisiología , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/tratamiento farmacológico , Tetrahidronaftalenos/farmacología , Animales , Dexametasona/toxicidad , Glucocorticoides/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/inducido químicamente , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Int Immunol ; 34(2): 81-95, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-34173833

RESUMEN

The heart is highly innervated by autonomic neurons, and dynamic autonomic regulation of the heart and blood vessels is essential for animals to carry out the normal activities of life. Cardiovascular diseases, including heart failure and myocardial infarction, are characterized in part by an imbalance in autonomic nervous system activation, with excess sympathetic and diminished parasympathetic activation. Notably, however, this is often accompanied by chronic inflammation within the cardiovascular tissues, which suggests there are interactions between autonomic dysregulation and inflammation. Recent studies have been unraveling the mechanistic links between autonomic nerves and immune cells within the cardiovascular system. The autonomic nervous system and immune system also act in concert to coordinate the actions of multiple organs that not only maintain homeostasis but also likely play key roles in disease-disease interactions, such as cardiorenal syndrome and multimorbidity. In this review, we summarize the physiological and pathological interactions between autonomic nerves and macrophages in the context of cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares , Animales , Sistema Nervioso Autónomo/fisiología , Corazón/inervación , Inflamación , Macrófagos
4.
Proc Natl Acad Sci U S A ; 117(25): 14365-14375, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513690

RESUMEN

Proper resolution of inflammation is vital for repair and restoration of homeostasis after tissue damage, and its dysregulation underlies various noncommunicable diseases, such as cardiovascular and metabolic diseases. Macrophages play diverse roles throughout initial inflammation, its resolution, and tissue repair. Differential metabolic reprogramming is reportedly required for induction and support of the various macrophage activation states. Here we show that a long noncoding RNA (lncRNA), lncFAO, contributes to inflammation resolution and tissue repair in mice by promoting fatty acid oxidation (FAO) in macrophages. lncFAO is induced late after lipopolysaccharide (LPS) stimulation of cultured macrophages and in Ly6Chi monocyte-derived macrophages in damaged tissue during the resolution and reparative phases. We found that lncFAO directly interacts with the HADHB subunit of mitochondrial trifunctional protein and activates FAO. lncFAO deletion impairs resolution of inflammation related to endotoxic shock and delays resolution of inflammation and tissue repair in a skin wound. These results demonstrate that by tuning mitochondrial metabolism, lncFAO acts as a node of immunometabolic control in macrophages during the resolution and repair phases of inflammation.


Asunto(s)
Ácidos Grasos/metabolismo , Inflamación/inmunología , Macrófagos/inmunología , Subunidad beta de la Proteína Trifuncional Mitocondrial/genética , ARN Largo no Codificante/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Lipopolisacáridos/inmunología , Activación de Macrófagos/genética , Macrófagos/metabolismo , Masculino , Ratones , Subunidad beta de la Proteína Trifuncional Mitocondrial/metabolismo , Oxidación-Reducción , Cultivo Primario de Células , ARN Largo no Codificante/genética , Piel/inmunología , Piel/lesiones , Cicatrización de Heridas/inmunología
5.
Proc Natl Acad Sci U S A ; 115(33): 8370-8375, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30061407

RESUMEN

The tumor suppressor p53 regulates multiple cellular functions, including energy metabolism. Metabolic deregulation is implicated in the pathogenesis of some cancers and in metabolic disorders and may result from the inactivation of p53 functions. Using RNA sequencing and ChIP sequencing of cancer cells and preadipocytes, we demonstrate that p53 modulates several metabolic processes via the transactivation of energy metabolism genes including dihydropyrimidinase-like 4 (DPYSL4). DPYSL4 is a member of the collapsin response mediator protein family, which is involved in cancer invasion and progression. Intriguingly, DPYSL4 overexpression in cancer cells and preadipocytes up-regulated ATP production and oxygen consumption, while DPYSL4 knockdown using siRNA or CRISPR/Cas9 down-regulated energy production. Furthermore, DPYSL4 was associated with mitochondrial supercomplexes, and deletion of its dihydropyrimidinase-like domain abolished its association and its ability to stimulate ATP production and suppress the cancer cell invasion. Mouse-xenograft and lung-metastasis models indicated that DPYSL4 expression compromised tumor growth and metastasis in vivo. Consistently, database analyses demonstrated that low DPYSL4 expression was significantly associated with poor survival of breast and ovarian cancers in accordance with its reduced expression in certain types of cancer tissues. Moreover, immunohistochemical analysis using the adipose tissue of obese patients revealed that DPYSL4 expression was positively correlated with INFg and body mass index in accordance with p53 activation. Together, these results suggest that DPYSL4 plays a key role in the tumor-suppressor function of p53 by regulating oxidative phosphorylation and the cellular energy supply via its association with mitochondrial supercomplexes, possibly linking to the pathophysiology of both cancer and obesity.


Asunto(s)
Adipocitos/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Neoplasias/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteína p53 Supresora de Tumor/fisiología , Adenosina Trifosfato/biosíntesis , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones SCID , Obesidad/metabolismo , Consumo de Oxígeno , Proteínas Supresoras de Tumor/fisiología
6.
Proc Natl Acad Sci U S A ; 115(23): 5980-5985, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784786

RESUMEN

The N6-methylation of internal adenosines (m6A) in mRNA has been quantified and localized throughout the transcriptome. However, the physiological significance of m6A in most highly methylated mRNAs is unknown. It was demonstrated previously that the circadian clock, based on transcription-translation negative feedback loops, is sensitive to the general inhibition of m6A. Here, we show that the Casein Kinase 1 Delta mRNA (Ck1δ), coding for a critical kinase in the control of circadian rhythms, cellular growth, and survival, is negatively regulated by m6A. Inhibition of Ck1δ mRNA methylation leads to increased translation of two alternatively spliced CK1δ isoforms, CK1δ1 and CK1δ2, uncharacterized until now. The expression ratio between these isoforms is tissue-specific, CK1δ1 and CK1δ2 have different kinase activities, and they cooperate in the phosphorylation of the circadian clock protein PER2. While CK1δ1 accelerates the circadian clock by promoting the decay of PER2 proteins, CK1δ2 slows it down by stabilizing PER2 via increased phosphorylation at a key residue on PER2 protein. These observations challenge the previously established model of PER2 phosphorylation and, given the multiple functions and targets of CK1δ, the existence of two isoforms calls for a re-evaluation of past research when CK1δ1 and CK1δ2 were simply CK1δ.


Asunto(s)
Quinasa Idelta de la Caseína/genética , Relojes Circadianos/genética , Metilación , Metiltransferasas/genética , ARN Mensajero/genética , Animales , Quinasa Idelta de la Caseína/metabolismo , Masculino , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas , Empalme del ARN/genética , ARN Mensajero/metabolismo
7.
Development ; 144(20): 3706-3718, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28870993

RESUMEN

The inner cell mass of the mouse blastocyst gives rise to the pluripotent epiblast (EPI), which forms the embryo proper, and the primitive endoderm (PrE), which forms extra-embryonic yolk sac tissues. All inner cells coexpress lineage markers such as Nanog and Gata6 at embryonic day (E) 3.25, and the EPI and PrE precursor cells eventually segregate to exclusively express Nanog and Gata6, respectively. Fibroblast growth factor (FGF)-extracellular signal-regulated kinase (ERK) signalling is involved in segregation of the EPI and PrE lineages; however, the mechanism involved in Fgf4 regulation is poorly understood. Here, we identified Klf5 as an upstream repressor of Fgf4Fgf4 was markedly upregulated in Klf5 knockout (KO) embryos at E3.0, and was downregulated in embryos overexpressing Klf5 Furthermore, Klf5 KO and overexpressing blastocysts showed skewed lineage specification phenotypes, similar to FGF4-treated preimplantation embryos and Fgf4 KO embryos, respectively. Inhibitors of the FGF receptor (Fgfr) and ERK pathways reversed the skewed lineage specification of Klf5 KO blastocysts. These data demonstrate that Klf5 suppresses Fgf4-Fgfr-ERK signalling, thus preventing precocious activation of the PrE specification programme.


Asunto(s)
Endodermo/metabolismo , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Blastocisto/metabolismo , Diferenciación Celular , Linaje de la Célula , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Células Madre Pluripotentes/citología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Factores de Tiempo
8.
Blood ; 132(23): 2470-2483, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30228234

RESUMEN

BCOR, encoding BCL-6 corepressor (BCOR), is X-linked and targeted by somatic mutations in various hematological malignancies including myelodysplastic syndrome (MDS). We previously reported that mice lacking Bcor exon 4 (Bcor ΔE4/y ) in the hematopoietic compartment developed NOTCH-dependent acute T-cell lymphoblastic leukemia (T-ALL). Here, we analyzed mice lacking Bcor exons 9 and 10 (Bcor ΔE9-10/y ), which express a carboxyl-terminal truncated BCOR that fails to interact with core effector components of polycomb repressive complex 1.1. Bcor ΔE9-10/y mice developed lethal T-ALL in a similar manner to Bcor ΔE4/y mice, whereas Bcor ΔE9-10/y hematopoietic cells showed a growth advantage in the myeloid compartment that was further enhanced by the concurrent deletion of Tet2 Tet2 Δ/Δ Bcor ΔE9-10/y mice developed lethal MDS with progressive anemia and leukocytopenia, inefficient hematopoiesis, and the morphological dysplasia of blood cells. Tet2 Δ/Δ Bcor ΔE9-10/y MDS cells reproduced MDS or evolved into lethal MDS/myeloproliferative neoplasms in secondary recipients. Transcriptional profiling revealed the derepression of myeloid regulator genes of the Cebp family and Hoxa cluster genes in Bcor ΔE9-10/y progenitor cells and the activation of p53 target genes specifically in MDS erythroblasts where massive apoptosis occurred. Our results reveal a tumor suppressor function of BCOR in myeloid malignancies and highlight the impact of Bcor insufficiency on the initiation and progression of MDS.


Asunto(s)
Secuencia de Aminoácidos , Exones , Síndromes Mielodisplásicos , Proteínas Represoras , Eliminación de Secuencia , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Int J Mol Sci ; 21(8)2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32295185

RESUMEN

Accumulation of lipid-laden (foam) cells in the arterial wall is known to be the earliest step in the pathogenesis of atherosclerosis. There is almost no doubt that atherogenic modified low-density lipoproteins (LDL) are the main sources of accumulating lipids in foam cells. Atherogenic modified LDL are taken up by arterial cells, such as macrophages, pericytes, and smooth muscle cells in an unregulated manner bypassing the LDL receptor. The present study was conducted to reveal possible common mechanisms in the interaction of macrophages with associates of modified LDL and non-lipid latex particles of a similar size. To determine regulatory pathways that are potentially responsible for cholesterol accumulation in human macrophages after the exposure to naturally occurring atherogenic or artificially modified LDL, we used transcriptome analysis. Previous studies of our group demonstrated that any type of LDL modification facilitates the self-association of lipoprotein particles. The size of such self-associates hinders their interaction with a specific LDL receptor. As a result, self-associates are taken up by nonspecific phagocytosis bypassing the LDL receptor. That is why we used latex beads as a stimulator of macrophage phagocytotic activity. We revealed at least 12 signaling pathways that were regulated by the interaction of macrophages with the multiple-modified atherogenic naturally occurring LDL and with latex beads in a similar manner. Therefore, modified LDL was shown to stimulate phagocytosis through the upregulation of certain genes. We have identified at least three genes (F2RL1, EIF2AK3, and IL15) encoding inflammatory molecules and associated with signaling pathways that were upregulated in response to the interaction of modified LDL with macrophages. Knockdown of two of these genes, EIF2AK3 and IL15, completely suppressed cholesterol accumulation in macrophages. Correspondingly, the upregulation of EIF2AK3 and IL15 promoted cholesterol accumulation. These data confirmed our hypothesis of the following chain of events in atherosclerosis: LDL particles undergo atherogenic modification; this is accompanied by the formation of self-associates; large LDL associates stimulate phagocytosis; as a result of phagocytosis stimulation, pro-inflammatory molecules are secreted; these molecules cause or at least contribute to the accumulation of intracellular cholesterol. This chain of events may explain the relationship between cholesterol accumulation and inflammation. The primary sequence of events in this chain is related to inflammatory response rather than cholesterol accumulation.


Asunto(s)
Colesterol/metabolismo , Células Espumosas/metabolismo , Metabolismo de los Lípidos , Transducción de Señal , Biomarcadores , Susceptibilidad a Enfermedades , Células Espumosas/patología , Perfilación de la Expresión Génica , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Modelos Biológicos
10.
Int J Mol Sci ; 21(3)2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-32012706

RESUMEN

Excessive accumulation of lipid inclusions in the arterial wall cells (foam cell formation) caused by modified low-density lipoprotein (LDL) is the earliest and most noticeable manifestation of atherosclerosis. The mechanisms of foam cell formation are not fully understood and can involve altered lipid uptake, impaired lipid metabolism, or both. Recently, we have identified the top 10 master regulators that were involved in the accumulation of cholesterol in cultured macrophages induced by the incubation with modified LDL. It was found that most of the identified master regulators were related to the regulation of the inflammatory immune response, but not to lipid metabolism. A possible explanation for this unexpected result is a stimulation of the phagocytic activity of macrophages by modified LDL particle associates that have a relatively large size. In the current study, we investigated gene regulation in macrophages using transcriptome analysis to test the hypothesis that the primary event occurring upon the interaction of modified LDL and macrophages is the stimulation of phagocytosis, which subsequently triggers the pro-inflammatory immune response. We identified genes that were up- or downregulated following the exposure of cultured cells to modified LDL or latex beads (inert phagocytosis stimulators). Most of the identified master regulators were involved in the innate immune response, and some of them were encoding major pro-inflammatory proteins. The obtained results indicated that pro-inflammatory response to phagocytosis stimulation precedes the accumulation of intracellular lipids and possibly contributes to the formation of foam cells. In this way, the currently recognized hypothesis that the accumulation of lipids triggers the pro-inflammatory response was not confirmed. Comparative analysis of master regulators revealed similarities in the genetic regulation of the interaction of macrophages with naturally occurring LDL and desialylated LDL. Oxidized and desialylated LDL affected a different spectrum of genes than naturally occurring LDL. These observations suggest that desialylation is the most important modification of LDL occurring in vivo. Thus, modified LDL caused the gene regulation characteristic of the stimulation of phagocytosis. Additionally, the knock-down effect of five master regulators, such as IL15, EIF2AK3, F2RL1, TSPYL2, and ANXA1, on intracellular lipid accumulation was tested. We knocked down these genes in primary macrophages derived from human monocytes. The addition of atherogenic naturally occurring LDL caused a significant accumulation of cholesterol in the control cells. The knock-down of the EIF2AK3 and IL15 genes completely prevented cholesterol accumulation in cultured macrophages. The knock-down of the ANXA1 gene caused a further decrease in cholesterol content in cultured macrophages. At the same time, knock-down of F2RL1 and TSPYL2 did not cause an effect. The results obtained allowed us to explain in which way the inflammatory response and the accumulation of cholesterol are related confirming our hypothesis of atherogenesis development based on the following viewpoints: LDL particles undergo atherogenic modifications that, in turn, accompanied by the formation of self-associates; large LDL associates stimulate phagocytosis; as a result of phagocytosis stimulation, pro-inflammatory molecules are secreted; these molecules cause or at least contribute to the accumulation of intracellular cholesterol. Therefore, it became obvious that the primary event in this sequence is not the accumulation of cholesterol but an inflammatory response.


Asunto(s)
Células Espumosas/metabolismo , Células Espumosas/patología , Lipoproteínas LDL/metabolismo , Fagocitosis , Biomarcadores , Células Espumosas/inmunología , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Metabolismo de los Lípidos , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Oxidación-Reducción , Fagocitosis/genética , Fagocitosis/inmunología , Transducción de Señal , Transcriptoma
11.
Int Immunol ; 30(11): 511-528, 2018 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-30165385

RESUMEN

Tissue injury triggers a complex series of cellular responses, starting from inflammation activated by tissue and cell damage and proceeding to healing. By clearing cell debris, activating and resolving inflammation and promoting fibrosis, macrophages play key roles in most, if not all, phases of the response to injury. Recent studies of the mechanisms underlying the initial inflammation and later tissue regeneration and repair revealed that macrophages bridge these processes in part by supporting and activating stem/progenitor cells, clearing damaged tissue, remodeling extracellular matrix to prepare scaffolding for regeneration and promoting angiogenesis. However, macrophages also have a central role in the development of pathology induced by failed resolution (e.g. chronic inflammation) and excessive scarring. In this review, we summarize the activities of macrophages in inflammation and healing in response to acute injury in tissues with differing regenerative capacities. While macrophages lead similar processes in response to tissue injury in these tissues, their priorities and the consequences of their activities differ among tissues. Moreover, the magnitude, nature and duration of injury also greatly affect cellular responses and healing processes. In particular, continuous injury and/or failed resolution of inflammation leads to chronic ailments in which macrophage activities may become detrimental.


Asunto(s)
Inflamación/inmunología , Inflamación/patología , Macrófagos/citología , Macrófagos/inmunología , Regeneración/inmunología , Cicatrización de Heridas/inmunología , Animales , Humanos , Macrófagos/patología
12.
J Pharmacol Sci ; 141(1): 56-63, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31611176

RESUMEN

Concomitant heart failure is associated with poor clinical outcome in dialysis patients. The arteriovenous shunt, created as vascular access for hemodialysis, increases ventricular volume-overload, predisposing patients to developing cardiac dysfunction. The integral function of mitochondrial respiration is critically important for the heart to cope with hemodynamic overload. The involvement, however, of mitochondrial activity or reactive oxygen species (ROS) in the pathogenesis of ventricular-overload-induced heart failure has not been fully elucidated. We herein report that disorganization of mitochondrial respiration increases mitochondrial ROS production in the volume-overloaded heart, leading to ventricular dysfunction. We adopted the murine arteriovenous fistula (AVF) model, which replicates the cardinal features of volume-overload-induced ventricular dysfunction. Enzymatic assays of cardiac mitochondria revealed that the activities of citrate synthase and NADH-quinone reductase (complex Ⅰ) were preserved in the AVF heart. In contrast, the activity of NADH oxidase supercomplex was significantly compromised, resulting in elevated ROS production. Importantly, the antioxidant N-acetylcysteine prevented the development of ventricular dilatation and cardiac dysfunction, suggesting a pathogenic role for ROS in dialysis-related cardiomyopathy. A cardioprotective effect was also observed in metformin-treated mice, illuminating its potential use in the management of heart failure complicating diabetic patients on dialysis.


Asunto(s)
Acetilcisteína/uso terapéutico , Antioxidantes/uso terapéutico , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/etiología , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Cardiomiopatías/metabolismo , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/prevención & control , Masculino , Ratones Endogámicos C57BL
13.
Int Heart J ; 60(2): 451-456, 2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30799373

RESUMEN

Chronic thromboembolic pulmonary hypertension (CTEPH) develops as a consequence of unresolved pulmonary embolism or clots in the pulmonary arteries. The obstruction not only reduces the area of the pulmonary vascular bed, but also elicits high pressure and high shear stress in the spared unobstructed arteries. Subsequent overflow of the small pulmonary arteries induces vascular remodeling, termed as overflow vasculopathy (OV). While the development of OV significantly contributes to the occurrence of pulmonary hypertension, its precise molecular mechanisms are yet to be determined.We established a novel murine pulmonary artery OV (PAOV) model, in which we resected left lung and induced redistribution of the cardiac output to the remaining pulmonary artery of the right lung. At 21 days after operation, mice showed an increase in the vascular media area, indicating the development of pulmonary arterial remodeling. In addition, right ventricular hypertrophy was detected in the PAOV model. Intriguingly, marked accumulation of F4/80-positive monocytes/macrophages was visualized in high-flow arteries, implying the role of an inflammatory process in the pathogenesis of overflow-induced vascular remodeling.


Asunto(s)
Hipertensión Pulmonar , Pulmón , Macrófagos/inmunología , Monocitos/inmunología , Remodelación Vascular/inmunología , Animales , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Pulmón/inmunología , Pulmón/patología , Ratones , Arteria Pulmonar/fisiopatología , Circulación Pulmonar/fisiología , Embolia Pulmonar/complicaciones
14.
Int Heart J ; 60(4): 958-963, 2019 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-31308330

RESUMEN

Myocardial infarction (MI) occurs when the heart muscle is severely damaged due to a decrease in blood flow from the coronary arteries. During recovery from an MI, cardiac fibroblasts become activated and produce extracellular matrices, contributing to the wound healing process in the damaged heart. Inappropriate activation of the fibroblasts leads to excessive fibrosis in the heart. However, the molecular pathways by which cardiac fibroblasts are activated have not yet been fully elucidated.Here we show that serum deprivation, which recapitulates the cellular microenvironment of the MI area, strikingly induces collagen production in C3H/10T1/2 cells. Based on transcriptomic and pharmacological studies, we found that cell cycle perturbation is directly linked to collagen production in fibroblasts. Importantly, collagen synthesis is increased independently of the transcriptional levels of type I collagen genes. These results reveal a novel mode of fibroblast activation in the ischemic area, which will allow us to gain insights into the molecular mechanisms underlying cardiac fibrosis and establish a basis for anti-fibrotic therapy.


Asunto(s)
Colágeno/biosíntesis , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Ciclo Celular , Células Cultivadas , Fibroblastos/metabolismo , Ratones , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Transducción de Señal
15.
Development ; 142(13): 2375-84, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26092847

RESUMEN

The protein CHD1 is a member of the family of ATPase-dependent chromatin remodeling factors. CHD1, which recognizes trimethylated histone H3 lysine 4, has been implicated in transcriptional activation in organisms ranging from yeast to humans. It is required for pre-mRNA maturation, maintenance of mouse embryonic stem cell pluripotency and rapid growth of the mouse epiblast. However, the function(s) of CHD1 in mouse preimplantation embryos has not yet been examined. Here, we show that loss of CHD1 function led to embryonic lethality after implantation. In mouse embryos in which Chd1 was targeted by siRNA microinjection, the expression of the key regulators of cell fate specification Pou5f1 (also known as Oct4), Nanog and Cdx2 was dramatically decreased, starting at mid-preimplantation gene activation (MGA). Moreover, expression of Hmgpi and Klf5, which regulate Pou5f1, Nanog and Cdx2, was also significantly suppressed at zygotic gene activation (ZGA). Suppression of Hmgpi expression in Chd1-knockdown embryos continued until the blastocyst stage, whereas suppression of Klf5 expression was relieved by the morula stage. Next, we rescued HMGPI expression via Hmgpi mRNA microinjection in Chd1-knockdown embryos. Consequently, Pou5f1, Nanog and Cdx2 expression was restored at MGA and live offspring were recovered. These findings indicate that CHD1 plays important roles in mouse early embryogenesis via activation of Hmgpi at ZGA.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario , Proteínas HMGB/metabolismo , Transducción de Señal , Animales , Proteínas de Unión al ADN/genética , Implantación del Embrión/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HMGB/genética , Humanos , Tamaño de la Camada , Ratones Endogámicos ICR , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/genética , Cigoto/metabolismo
16.
Microsurgery ; 37(8): 910-916, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28621805

RESUMEN

BACKGROUND: Although early detection is valuable for secondary lymphedema treatment, existing screening tests are not popular. This study aimed to propose a novel method of screening lymphedema patients based on the thickness of the subcutaneous fat measured with perioperative computed tomography (CT) and present the results from evaluation of patients who underwent those examinations was performed. METHOD: The medical records of 96 gynecological cancer patients and 189 breast cancer patients, whose lymphatic function was assessed with indocyanine green lymphography, were reviewed. In gynecological cancer patients, the perioperative temporal subcutaneous fat thickness index (T-SFTI) was calculated from presurgical and follow-up CT data, and in breast cancer patients, the postoperative crosswise subcutaneous fat thickness index (C-SFTI) was calculated. In lower extremity lymphedema patients, the effect of lymphaticovenular anastomosis (LVA) was also evaluated using T-SFTI. RESULTS: Perioperative T-SFTI was assessed in 180 lower extremities, and it was significantly higher in 46 lymphatic dysfunction limbs (1.21 ± 0.08) than in 134 normal lymphatic function limbs (1.03 ± 0.08), (P < .01). Postoperative C-SFTI was assessed in 53 upper extremity, and it was significantly higher in 11 lymphatic dysfunction limbs (1.31 ± 0.21) than in 42 normal lymphatic function limbs (1.01 ± 0.06), (P < .01). In lower extremity lymphedema patients, T-SFTI improved significantly after planned conservative treatments and LVA (P = .04). CONCLUSION: Assessment of subcutaneous fat thickness using CT is useful for screening early stage lymphedema. If the efficacy of this method is validated, patients worldwide may be assessed using the same criterion.


Asunto(s)
Neoplasias de la Mama/cirugía , Neoplasias de los Genitales Femeninos/cirugía , Linfedema/diagnóstico por imagen , Complicaciones Posoperatorias/diagnóstico por imagen , Grasa Subcutánea/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Anastomosis Quirúrgica , Colorantes , Femenino , Humanos , Verde de Indocianina , Extremidad Inferior , Linfedema/etiología , Linfografía , Persona de Mediana Edad , Complicaciones Posoperatorias/etiología , Curva ROC , Estudios Retrospectivos , Extremidad Superior
17.
Clin Calcium ; 27(6): 795-801, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28536316

RESUMEN

Adipose tissues are the major organ that controls systemic energy metabolism and maintain homeostasis by storing lipids, dissipating them as heat, and producing various adipokines. There are two major classes of adipocytes: white and brown adipocytes. White adipocytes store and release lipids, while brown adipocytes burn substrates to produce heat. In addition to classical brown adipose tissues consisting of brown adipocytes, cold exposure and ß3 stimulation induce development of brown cell-like "beige" adipocytes in white adipose tissues. There appear to be multiple adipocyte progenitor cell populations of different developmental origins. In this article, we overview white and brown/beige adipocyte differentiation in development and obesity. Adipocytes differentiate in complex interplays with various stromal cells, including vascular, immune and neuronal cells. Elucidation of the cellular interplays would lead to identification of novel therapeutic targets for obesity and metabolic syndrome.


Asunto(s)
Adipocitos Marrones/citología , Adipocitos Blancos/citología , Diferenciación Celular , Células Madre/citología , Animales , Comunicación Celular , Humanos
18.
Am J Physiol Lung Cell Mol Physiol ; 311(4): L704-L713, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27542805

RESUMEN

Angiopoietin-like protein 2 (ANGPTL2) is a chronic inflammatory mediator that, when deregulated, is associated with various pathologies. However, little is known about its activity in lung. To assess a possible lung function, we generated a rabbit monoclonal antibody that specifically recognizes mouse ANGPTL2 and then evaluated protein expression in mouse lung tissue. We observed abundant ANGPTL2 expression in both alveolar epithelial type I and type II cells and in resident alveolar macrophages under normal conditions. To assess ANGPTL2 function, we compared lung phenotypes in Angptl2 knockout (KO) and wild-type mice but observed no overt changes. We then generated a bleomycin-induced interstitial pneumonia model using wild-type and Angptl2 KO mice. Bleomycin-treated wild-type mice showed specifically upregulated ANGPTL2 expression in areas of severe fibrosing interstitial pneumonia, while Angptl2 KO mice developed more severe lung fibrosis than did comparably treated wild-type mice. Lung fibrosis seen following bone marrow transplant was comparable in wild-type or Angptl2 KO mice treated with bleomycin, suggesting that Angptl2 loss in myeloid cells does not underlie fibrotic phenotypes. We conclude that Angptl2 deficiency in lung epithelial cells and resident alveolar macrophages causes severe lung fibrosis seen following bleomycin treatment, suggesting that ANGPTL2 derived from these cell types plays a protective role against fibrosis in lung.


Asunto(s)
Angiopoyetinas/genética , Enfermedades Pulmonares Intersticiales/genética , Fibrosis Pulmonar/genética , Células 3T3-L1 , Células Epiteliales Alveolares/metabolismo , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/metabolismo , Animales , Bleomicina , Pulmón/patología , Enfermedades Pulmonares Intersticiales/inducido químicamente , Enfermedades Pulmonares Intersticiales/patología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Trombospondina 1/genética , Trombospondina 1/metabolismo
19.
Clin Exp Pharmacol Physiol ; 43(3): 294-303, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26728525

RESUMEN

Obesity is known to associate with low-grade, sustained, systemic inflammation, which is considered to be a key pathological basis for obesity-associated diseases such as diabetes and atherosclerosis. Immune cells, including both lymphocytes and macrophages, play physiological and pathological roles in adipose tissue. They increasingly infiltrate obese adipose tissue as body weight is gained, after which the infiltrated cells promote adipose tissue inflammation and strongly impact systemic metabolism. Recent studies have shown that the immune and metabolic systems are highly integrated with one another. This recognition has provided new insight into the mechanisms of metabolic diseases. In addition to the link at the tissue level, studies have shown that immune cell function is coordinately regulated with cellular metabolism. This review summarizes the current understanding of the specific metabolic signatures adopted by lymphocytes and macrophages to mediate proper effecter function. These findings will be related to the regulation of adipose tissue homeostasis and inflammation.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Animales , Proliferación Celular , Regulación de la Expresión Génica , Glucólisis , Humanos , Metabolismo de los Lípidos , Macrófagos/citología , Macrófagos/inmunología
20.
Nihon Rinsho ; 74(9): 1441-1445, 2016 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-30557474

RESUMEN

Chronic inflammation is the common pathological basis for age-associated diseases. Chronic activation of basic inflammatory sates is known to associate with aging. Changes in immune system (immunosenescence), tissue microenvironment, such as the accumulation of cell debris, and systemic changes in metabolic and hormonal signals, likely contribute to the development of chronic inflammation. Inflammaging is coined to indicate the close link between aging and inflammation. In this review we will address how age-associated changes in body promote chronic inflammation.


Asunto(s)
Envejecimiento , Mediadores de Inflamación , Inflamación , Envejecimiento/inmunología , Humanos , Sistema Inmunológico , Mediadores de Inflamación/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA