Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Biol Chem ; 299(12): 105426, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37926281

RESUMEN

S-palmitoylation is a reversible lipid modification catalyzed by 23 S-acyltransferases with a conserved zinc finger aspartate-histidine-histidine-cysteine (zDHHC) domain that facilitates targeting of proteins to specific intracellular membranes. Here we performed a gain-of-function screen in the mouse and identified the Golgi-localized enzymes zDHHC3 and zDHHC7 as regulators of cardiac hypertrophy. Cardiomyocyte-specific transgenic mice overexpressing zDHHC3 show cardiac disease, and S-acyl proteomics identified the small GTPase Rac1 as a novel substrate of zDHHC3. Notably, cardiomyopathy and congestive heart failure in zDHHC3 transgenic mice is preceded by enhanced Rac1 S-palmitoylation, membrane localization, activity, downstream hypertrophic signaling, and concomitant induction of all Rho family small GTPases whereas mice overexpressing an enzymatically dead zDHHC3 mutant show no discernible effect. However, loss of Rac1 or other identified zDHHC3 targets Gαq/11 or galectin-1 does not diminish zDHHC3-induced cardiomyopathy, suggesting multiple effectors and pathways promoting decompensation with sustained zDHHC3 activity. Genetic deletion of Zdhhc3 in combination with Zdhhc7 reduces cardiac hypertrophy during the early response to pressure overload stimulation but not over longer time periods. Indeed, cardiac hypertrophy in response to 2 weeks of angiotensin-II infusion is not diminished by Zdhhc3/7 deletion, again suggesting other S-acyltransferases or signaling mechanisms compensate to promote hypertrophic signaling. Taken together, these data indicate that the activity of zDHHC3 and zDHHC7 at the cardiomyocyte Golgi promote Rac1 signaling and maladaptive cardiac remodeling, but redundant signaling effectors compensate to maintain cardiac hypertrophy with sustained pathological stimulation in the absence of zDHHC3/7.


Asunto(s)
Cardiomiopatías , Miocitos Cardíacos , Animales , Ratones , Aciltransferasas/genética , Aciltransferasas/metabolismo , Cardiomegalia/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Histidina/metabolismo , Lipoilación , Ratones Transgénicos , Miocitos Cardíacos/metabolismo
2.
Cells ; 12(17)2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37681905

RESUMEN

RATIONALE: The adult cardiac extracellular matrix (ECM) is largely comprised of type I collagen. In addition to serving as the primary structural support component of the cardiac ECM, type I collagen also provides an organizational platform for other ECM proteins, matricellular proteins, and signaling components that impact cellular stress sensing in vivo. OBJECTIVE: Here we investigated how the content and integrity of type I collagen affect cardiac structure function and response to injury. METHODS AND RESULTS: We generated and characterized Col1a2-/- mice using standard gene targeting. Col1a2-/- mice were viable, although by young adulthood their hearts showed alterations in ECM mechanical properties, as well as an unanticipated activation of cardiac fibroblasts and induction of a progressive fibrotic response. This included augmented TGFß activity, increases in fibroblast number, and progressive cardiac hypertrophy, with reduced functional performance by 9 months of age. Col1a2-loxP-targeted mice were also generated and crossed with the tamoxifen-inducible Postn-MerCreMer mice to delete the Col1a2 gene in myofibroblasts with pressure overload injury. Interestingly, while germline Col1a2-/- mice showed gradual pathologic hypertrophy and fibrosis with aging, the acute deletion of Col1a2 from activated adult myofibroblasts showed a loss of total collagen deposition with acute cardiac injury and an acute reduction in pressure overload-induce cardiac hypertrophy. However, this reduction in hypertrophy due to myofibroblast-specific Col1a2 deletion was lost after 2 and 6 weeks of pressure overload, as fibrotic deposition accumulated. CONCLUSIONS: Defective type I collagen in the heart alters the structural integrity of the ECM and leads to cardiomyopathy in adulthood, with fibroblast expansion, activation, and alternate fibrotic ECM deposition. However, acute inhibition of type I collagen production can have an anti-fibrotic and anti-hypertrophic effect.


Asunto(s)
Cardiomiopatías , Colágeno Tipo I , Animales , Ratones , Cardiomegalia/genética , Colágeno Tipo I/genética , Fibrosis
3.
Proc Natl Acad Sci U S A ; 120(19): e2213696120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126682

RESUMEN

To better understand the genetic basis of heart disease, we identified a variant in the Flightless-I homolog (FLII) gene that generates a R1243H missense change and predisposes to cardiac remodeling across multiple previous human genome-wide association studies (GWAS). Since this gene is of unknown function in the mammalian heart we generated gain- and loss-of-function genetically altered mice, as well as knock-in mice with the syntenic R1245H amino acid substitution, which showed that Flii protein binds the sarcomeric actin thin filament and influences its length. Deletion of Flii from the heart, or mice with the R1245H amino acid substitution, show cardiomyopathy due to shortening of the actin thin filaments. Mechanistically, Flii is a known actin binding protein that we show associates with tropomodulin-1 (TMOD1) to regulate sarcomere thin filament length. Indeed, overexpression of leiomodin-2 in the heart, which lengthens the actin-containing thin filaments, partially rescued disease due to heart-specific deletion of Flii. Collectively, the identified FLII human variant likely increases cardiomyopathy risk through an alteration in sarcomere structure and associated contractile dynamics, like other sarcomere gene-based familial cardiomyopathies.


Asunto(s)
Actinas , Cardiomiopatías , Humanos , Animales , Ratones , Actinas/metabolismo , Sarcómeros/metabolismo , Estudio de Asociación del Genoma Completo , Citoesqueleto de Actina/metabolismo , Cardiomiopatías/metabolismo , Mamíferos/genética , Proteínas de Microfilamentos/metabolismo , Transactivadores/metabolismo , Tropomodulina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Musculares/metabolismo
4.
Front Physiol ; 14: 1054169, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36733907

RESUMEN

Introduction: The ribosomal protein L3-like (RPL3L) is a heart and skeletal muscle-specific ribosomal protein and paralogue of the more ubiquitously expressed RPL3 protein. Mutations in the human RPL3L gene are linked to childhood cardiomyopathy and age-related atrial fibrillation, yet the function of RPL3L in the mammalian heart remains unknown. Methods and Results: Here, we observed that mouse cardiac ventricles express RPL3 at birth, where it is gradually replaced by RPL3L in adulthood but re-expressed with induction of hypertrophy in adults. Rpl3l gene-deleted mice were generated to examine the role of this gene in the heart, although Rpl3l -/- mice showed no overt changes in cardiac structure or function at baseline or after pressure overload hypertrophy, likely because RPL3 expression was upregulated and maintained in adulthood. mRNA expression analysis and ribosome profiling failed to show differences between the hearts of Rpl3l null and wild type mice in adulthood. Moreover, ribosomes lacking RPL3L showed no differences in localization within cardiomyocytes compared to wild type controls, nor was there an alteration in cardiac tissue ultrastructure or mitochondrial function in adult Rpl3l -/- mice. Similarly, overexpression of either RPL3 or RPL3L with adeno-associated virus -9 in the hearts of mice did not cause discernable pathology. However, by 18 months of age Rpl3l -/- null mice had significantly smaller hearts compared to wild type littermates. Conclusion: Thus, deletion of Rpl3l forces maintenance of RPL3 expression within the heart that appears to fully compensate for the loss of RPL3L, although older Rpl3l -/- mice showed a mild but significant reduction in heart weight.

5.
Nat Commun ; 12(1): 843, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594062

RESUMEN

Adaptive thermogenesis is essential for survival, and therefore is tightly regulated by a central neural circuit. Here, we show that microRNA (miR)-33 in the brain is indispensable for adaptive thermogenesis. Cold stress increases miR-33 levels in the hypothalamus and miR-33-/- mice are unable to maintain body temperature in cold environments due to reduced sympathetic nerve activity and impaired brown adipose tissue (BAT) thermogenesis. Analysis of miR-33f/f dopamine-ß-hydroxylase (DBH)-Cre mice indicates the importance of miR-33 in Dbh-positive cells. Mechanistically, miR-33 deficiency upregulates gamma-aminobutyric acid (GABA)A receptor subunit genes such as Gabrb2 and Gabra4. Knock-down of these genes in Dbh-positive neurons rescues the impaired cold-induced thermogenesis in miR-33f/f DBH-Cre mice. Conversely, increased gene dosage of miR-33 in mice enhances thermogenesis. Thus, miR-33 in the brain contributes to maintenance of BAT thermogenesis and whole-body metabolism via enhanced sympathetic nerve tone through suppressing GABAergic inhibitory neurotransmission. This miR-33-mediated neural mechanism may serve as a physiological adaptive defense mechanism for several stresses including cold stress.


Asunto(s)
MicroARNs/metabolismo , Sistema Nervioso Simpático/fisiología , Termogénesis/genética , Tejido Adiposo Pardo/fisiología , Animales , Temperatura Corporal/fisiología , Peso Corporal , Encéfalo/metabolismo , Línea Celular , Frío , Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Humanos , Integrasas/metabolismo , Masculino , Ratones , Ratones Obesos , MicroARNs/genética , Consumo de Oxígeno/fisiología , Fenotipo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
6.
Commun Biol ; 3(1): 434, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792557

RESUMEN

Recent high-throughput approaches have revealed a vast number of transcripts with unknown functions. Many of these transcripts are long noncoding RNAs (lncRNAs), and intergenic region-derived lncRNAs are classified as long intergenic noncoding RNAs (lincRNAs). Although Myosin heavy chain 6 (Myh6) encoding primary contractile protein is down-regulated in stressed hearts, the underlying mechanisms are not fully clarified especially in terms of lincRNAs. Here, we screen upregulated lincRNAs in pressure overloaded hearts and identify a muscle-abundant lincRNA termed Lionheart. Compared with controls, deletion of the Lionheart in mice leads to decreased systolic function and a reduction in MYH6 protein levels following pressure overload. We reveal decreased MYH6 results from an interaction between Lionheart and Purine-rich element-binding protein A after pressure overload. Furthermore, human LIONHEART levels in left ventricular biopsy specimens positively correlate with cardiac systolic function. Our results demonstrate Lionheart plays a pivotal role in cardiac remodeling via regulation of MYH6.


Asunto(s)
Corazón/fisiopatología , Presión , ARN Largo no Codificante/genética , Sístole/genética , Animales , Biopsia , Dependovirus/metabolismo , Ventrículos Cardíacos/ultraestructura , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/metabolismo , Ratas , Regulación hacia Arriba/genética
7.
JACC Basic Transl Sci ; 4(6): 701-714, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31709319

RESUMEN

No effective treatment is yet available to reduce infarct size and improve clinical outcomes after acute myocardial infarction by enhancing early reperfusion therapy using primary percutaneous coronary intervention. The study showed that Kyoto University Substance 121 (KUS121) reduced endoplasmic reticulum stress, maintained adenosine triphosphate levels, and ameliorated the infarct size in a murine cardiac ischemia and reperfusion injury model. The study confirmed the cardioprotective effect of KUS121 in a porcine ischemia and reperfusion injury model. These findings confirmed that KUS121 is a promising novel therapeutic agent for myocardial infarction in conjunction with primary percutaneous coronary intervention.

8.
J Am Heart Assoc ; 8(13): e012609, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31242815

RESUMEN

Background Micro RNA (miR)-33 targets cholesterol transporter ATP -binding cassette protein A1 and other antiatherogenic targets and contributes to atherogenic progression. Its inhibition or deletion is known to result in the amelioration of atherosclerosis in mice. However, mice lack the other member of the miR-33 family, miR-33b, which exists in humans and other large mammals. Thus, precise evaluation and comparison of the responsibilities of these 2 miRs during the progression of atherosclerosis has not been reported, although they are essential. Methods and Results In this study, we performed a comprehensive analysis of the difference between the function of miR-33a and miR-33b using genetically modified mice. We generated 4 strains with or without miR-33a and miR-33b. Comparison between mice with only miR-33a (wild-type mice) and mice with only miR-33b (miR-33a-/-/miR-33b+/+) revealed the dominant expression of miR-33b in the liver. To evaluate the whole body atherogenic potency of miR-33a and miR-33b, we developed apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice and apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice. With a high-fat and high-cholesterol diet, the apolipoprotein E-deficient/miR-33a-/-/miR-33b+/+ mice developed increased atherosclerotic plaque versus apolipoprotein E-deficient/miR-33a+/+/miR-33b-/- mice, in line with the predominant expression of miR-33b in the liver and worsened serum cholesterol profile. By contrast, a bone marrow transplantation study showed no significant difference, which was consistent with the relevant expression levels of miR-33a and miR-33b in bone marrow cells. Conclusions The miR-33 family exhibits differences in distribution and regulation and particularly in the progression of atherosclerosis; miR-33b would be more potent than miR-33a.


Asunto(s)
Aterosclerosis/genética , Hepatocitos/metabolismo , Hígado/metabolismo , MicroARNs/genética , Placa Aterosclerótica/genética , Animales , Apolipoproteínas B/metabolismo , Trasplante de Médula Ósea , Colesterol/metabolismo , Colesterol en la Dieta , Dieta Alta en Grasa , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Ratones Noqueados para ApoE , Ratones Transgénicos , MicroARNs/metabolismo , Triglicéridos/metabolismo
9.
Clin Sci (Lond) ; 133(4): 583-595, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30777884

RESUMEN

Recent reports, including ours, have indicated that microRNA (miR)-33 located within the intron of sterol regulatory element binding protein (SREBP) 2 controls cholesterol homeostasis and can be a potential therapeutic target for the treatment of atherosclerosis. Here, we show that SPAST, which encodes a microtubule-severing protein called SPASTIN, was a novel target gene of miR-33 in human. Actually, the miR-33 binding site in the SPAST 3'-UTR is conserved not in mice but in mid to large mammals, and it is impossible to clarify the role of miR-33 on SPAST in mice. We demonstrated that inhibition of miR-33a, a major form of miR-33 in human neurons, via locked nucleic acid (LNA)-anti-miR ameliorated the pathological phenotype in hereditary spastic paraplegia (HSP)-SPG4 patient induced pluripotent stem cell (iPSC)-derived cortical neurons. Thus, miR-33a can be a potential therapeutic target for the treatment of HSP-SPG4.


Asunto(s)
Terapia Genética/métodos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Oligonucleótidos/genética , Paraplejía Espástica Hereditaria/terapia , Espastina/genética , Regiones no Traducidas 3' , Sitios de Unión , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Células-Madre Neurales/patología , Neuritas/patología , Neurogénesis , Oligonucleótidos/metabolismo , Fenotipo , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo , Paraplejía Espástica Hereditaria/patología , Espastina/metabolismo
10.
Sci Rep ; 8(1): 16749, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30425314

RESUMEN

Acute cardiac rupture and adverse left ventricular (LV) remodeling causing heart failure are serious complications of acute myocardial infarction (MI). While cardio-hepatic interactions have been recognized, their role in MI remains unknown. We treated cultured cardiomyocytes with conditioned media from various cell types and analyzed the media by mass spectrometry to identify α1-microglobulin (AM) as an Akt-activating hepatokine. In mouse MI model, AM protein transiently distributed in the infarct and border zones during the acute phase, reflecting infiltration of AM-bound macrophages. AM stimulation activated Akt, NFκB, and ERK signaling and enhanced inflammation as well as macrophage migration and polarization, while inhibited fibrogenesis-related mRNA expression in cultured macrophages and cardiac fibroblasts. Intramyocardial AM administration exacerbated macrophage infiltration, inflammation, and matrix metalloproteinase 9 mRNA expression in the infarct and border zones, whereas disturbed fibrotic repair, then provoked acute cardiac rupture in MI. Shotgun proteomics and lipid pull-down analysis found that AM partly binds to phosphatidic acid (PA) for its signaling and function. Furthermore, systemic delivery of a selective inhibitor of diacylglycerol kinase α-mediated PA synthesis notably reduced macrophage infiltration, inflammation, matrix metalloproteinase activity, and adverse LV remodeling in MI. Therefore, targeting AM signaling could be a novel pharmacological option to mitigate adverse LV remodeling in MI.


Asunto(s)
alfa-Globulinas/metabolismo , Hormonas/metabolismo , Infarto del Miocardio/patología , Transducción de Señal , Animales , Membrana Celular/metabolismo , Movimiento Celular , Activación Enzimática , Fibrosis , Inflamación/metabolismo , Hígado/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ácidos Fosfatidicos/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remodelación Ventricular
11.
Arterioscler Thromb Vasc Biol ; 38(10): 2460-2473, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354203

RESUMEN

Objective- Atherosclerosis is a common disease caused by a variety of metabolic and inflammatory disturbances. MicroRNA (miR)-33a within SREBF2 (sterol regulatory element-binding factor 2) is a potent target for treatment of atherosclerosis through regulating both aspects; however, the involvement of miR-33b within SREBF1 remains largely unknown. Although their host genes difference could lead to functional divergence of miR-33a/b, we cannot dissect the roles of miR-33a/b in vivo because of lack of miR-33b sequences in mice, unlike human. Approach and Results- Here, we analyzed the development of atherosclerosis using miR-33b knock-in humanized mice under apolipoprotein E-deficient background. MiR-33b is prominent both in human and mice on atheroprone condition. MiR-33b reduced serum high-density lipoprotein cholesterol levels and systemic reverse cholesterol transport. MiR-33b knock-in macrophages showed less cholesterol efflux capacity and higher inflammatory state via regulating lipid rafts. Thus, miR-33b promotes vulnerable atherosclerotic plaque formation. Furthermore, bone marrow transplantation experiments strengthen proatherogenic roles of macrophage miR-33b. Conclusions- Our data demonstrated critical roles of SREBF1-miR-33b axis on both lipid profiles and macrophage phenotype remodeling and indicate that miR-33b is a promising target for treating atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , MicroARNs/metabolismo , Placa Aterosclerótica , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Trasplante de Médula Ósea , Estudios de Casos y Controles , HDL-Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Absorción Intestinal , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , MicroARNs/genética , Persona de Mediana Edad , Fenotipo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Triglicéridos/sangre
12.
Sci Rep ; 8(1): 8553, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29867212

RESUMEN

Recent evidence suggests that the accumulation of macrophages as a result of obesity-induced adipose tissue hypoxia is crucial for the regulation of tissue fibrosis, but the molecular mechanisms underlying adipose tissue fibrosis are still unknown. In this study, we revealed that periostin (Postn) is produced at extraordinary levels by adipose tissue after feeding with a high-fat diet (HFD). Postn was secreted at least from macrophages in visceral adipose tissue during the development of obesity, possibly due to hypoxia. Postn-/- mice had lower levels of crown-like structure formation and fibrosis in adipose tissue and were protected from liver steatosis. These mice also showed amelioration in systemic insulin resistance compared with HFD-fed WT littermates. Mice deficient in Postn in their hematopoietic compartment also had lower levels of inflammation in adipose tissue, in parallel with a reduction in ectopic lipid accumulation compared with the controls. Our data indicated that the regulation of Postn in visceral fat could be beneficial for the maintenance of healthy adipose tissue in obesity.


Asunto(s)
Moléculas de Adhesión Celular/deficiencia , Celulitis (Flemón)/metabolismo , Grasas de la Dieta/efectos adversos , Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Animales , Celulitis (Flemón)/inducido químicamente , Celulitis (Flemón)/genética , Celulitis (Flemón)/patología , Grasas de la Dieta/farmacología , Fibrosis , Grasa Intraabdominal/patología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología
13.
Mol Cell Biol ; 38(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712758

RESUMEN

MicroRNA 33 (miR-33) targets ATP-binding cassette transporter A1 (ABCA1), and its deficiency increases serum high-density lipoprotein (HDL)-cholesterol (HDL-C) and ameliorates atherosclerosis. Although we previously reported that miR-33 deficiency increased peripheral Ly6Chigh monocytes on an ApoE-deficient background, the effect of miR-33 on the monocyte population has not been fully elucidated, especially in a wild-type (WT) background. We found that Ly6Chigh monocytes in miR-33-/- mice were decreased in peripheral blood and increased in bone marrow (BM). Expansion of myeloid progenitors and decreased apoptosis in Lin- Sca1+ c-Kit+ (LSK) cells were observed in miR-33-/- mice. A BM transplantation study and competitive repopulation assay revealed that hematopoietic miR-33 deficiency caused myeloid expansion and increased peripheral Ly6Chigh monocytes and that nonhematopoietic miR-33 deficiency caused reduced peripheral Ly6Chigh monocytes. Expression of high-mobility group AT-hook 2 (HMGA2) targeted by miR-33 increased in miR-33-deficient LSK cells, and its knockdown abolished the reduction of apoptosis. Transduction of human apolipoprotein A1 and ABCA1 in WT mouse liver increased HDL-C and reduced peripheral Ly6Chigh monocytes. These data indicate that miR-33 deficiency affects distribution of inflammatory monocytes through dual pathways. One pathway involves the enhancement of Hmga2 expression in hematopoietic stem cells to increase Ly6Chigh monocytes, and the other involves the elevation of HDL-C to decrease peripheral Ly6Chigh monocytes.


Asunto(s)
Antígenos Ly/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Monocitos/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Apolipoproteínas E/metabolismo , Apoptosis , Aterosclerosis/genética , Aterosclerosis/metabolismo , HDL-Colesterol/sangre , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Monocitos/clasificación , Monocitos/citología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética
14.
Circ J ; 82(5): 1231-1236, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29526985

RESUMEN

Protein-coding genes account for less than 2% of the whole genome. However, the advances in RNA sequencing and genome-wide analysis have demonstrated that most of the genome is capable of being transcribed. Moreover, recent studies have suggested that long non-coding RNAs (lncRNAs) are critical regulators of gene expression and epigenesis in both physiological and disease states. Several lncRNAs are functionally involved in cardiovascular diseases and may be potential therapeutic targets. Here, we review the current strategies for the discovery of functional lncRNAs and recently discovered lncRNAs in the cardiovascular field, focusing on cardiac development, hypertrophy, heart failure, and atherosclerosis. We also discuss the therapeutic potentials of synthetic RNAs to modulate these lncRNAs and future directions in this research field.


Asunto(s)
Aterosclerosis , Cardiomegalia , Insuficiencia Cardíaca , ARN Largo no Codificante , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
15.
JACC Cardiovasc Interv ; 11(2): 145-157, 2018 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-29289632

RESUMEN

OBJECTIVES: The aim of this study was to evaluate the prognostic impact of left ventricular ejection fraction (LVEF) in patients with severe aortic stenosis (AS). BACKGROUND: The prognostic impact of LVEF in severe AS remains controversial. METHODS: Among 3,815 consecutive patients with severe AS enrolled in the CURRENT AS (Contemporary Outcomes After Surgery and Medical Treatment in Patients With Severe Aortic Stenosis) registry, the present study population consisted of 3,794 patients after excluding 21 patients without LVEF data. Patients were divided into 4 groups according to LVEF at index echocardiography (<50%, 50% to 59%, 60% to 69%, and ≥70%; conservative strategy: n = 388, n = 390, n = 1,025, and n = 800; initial aortic valve replacement strategy: n = 206, n = 170, n = 375, and n = 440). Echocardiographic data were site reported, and there was no echocardiography core laboratory. RESULTS: In the conservative group, the cumulative 5-year incidence of the primary outcome measure (a composite of aortic valve-related death or heart failure hospitalization) was significantly higher in patients with LVEFs <50% and 50% to 59% than in those with LVEFs 60% to 69% and ≥70% (72.3%, 58.4%, 38.7%, and 35.0%, respectively, p < 0.001), whereas in the initial aortic valve replacement group, the negative effect of low LVEF was markedly attenuated (20.2%, 20.3%, 17.7%, and 12.4%, respectively, p = 0.03). After adjusting for confounders, LVEF <50% (hazard ratio: 1.82; 95% confidence interval: 1.44 to 2.28; p < 0.001) and 50% to 59% (hazard ratio: 1.77; 95% confidence interval: 1.42 to 2.20; p < 0.001) but not 60% to 69% (hazard ratio: 1.14; 95% confidence interval: 0.94 to 1.39; p = 0.17) were independently associated with poorer outcomes compared with LVEF ≥70% (reference) in the conservative group. In the initial aortic valve replacement group, the adjusted risk for the primary outcome measure was not significantly different across the 4 LVEF groups. CONCLUSIONS: This study demonstrates that survival in patients with severe AS is impaired when LVEF is <60%, and these findings have implications for decision making with regard to the timing of surgical intervention.


Asunto(s)
Estenosis de la Válvula Aórtica/fisiopatología , Volumen Sistólico , Función Ventricular Izquierda , Anciano , Anciano de 80 o más Años , Estenosis de la Válvula Aórtica/diagnóstico por imagen , Estenosis de la Válvula Aórtica/mortalidad , Estenosis de la Válvula Aórtica/cirugía , Tratamiento Conservador/efectos adversos , Tratamiento Conservador/mortalidad , Progresión de la Enfermedad , Ecocardiografía Doppler , Femenino , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Implantación de Prótesis de Válvulas Cardíacas/mortalidad , Hospitalización , Humanos , Japón , Masculino , Sistema de Registros , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Resultado del Tratamiento
16.
Arterioscler Thromb Vasc Biol ; 37(11): 2161-2170, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28882868

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is an increasingly prevalent and ultimately fatal disease with no effective pharmacological treatment. Because matrix degradation induced by vascular inflammation is the major pathophysiology of AAA, attenuation of this inflammation may improve its outcome. Previous studies suggested that miR-33 (microRNA-33) inhibition and genetic ablation of miR-33 increased serum high-density lipoprotein cholesterol and attenuated atherosclerosis. APPROACH AND RESULTS: MiR-33a-5p expression in central zone of human AAA was higher than marginal zone. MiR-33 deletion attenuated AAA formation in both mouse models of angiotensin II- and calcium chloride-induced AAA. Reduced macrophage accumulation and monocyte chemotactic protein-1 expression were observed in calcium chloride-induced AAA walls in miR-33-/- mice. In vitro experiments revealed that peritoneal macrophages from miR-33-/- mice showed reduced matrix metalloproteinase 9 expression levels via c-Jun N-terminal kinase inactivation. Primary aortic vascular smooth muscle cells from miR-33-/- mice showed reduced monocyte chemotactic protein-1 expression by p38 mitogen-activated protein kinase attenuation. Both of the inactivation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were possibly because of the increase of ATP-binding cassette transporter A1 that is a well-known target of miR-33. Moreover, high-density lipoprotein cholesterol derived from miR-33-/- mice reduced expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells. Bone marrow transplantation experiments indicated that miR-33-deficient bone marrow cells ameliorated AAA formation in wild-type recipients. MiR-33 deficiency in recipient mice was also shown to contribute the inhibition of AAA formation. CONCLUSIONS: These data strongly suggest that inhibition of miR-33 will be effective as a novel strategy for treating AAA.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/prevención & control , Aortitis/prevención & control , Mediadores de Inflamación/metabolismo , MicroARNs/metabolismo , Angiotensina II , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Aortitis/inducido químicamente , Aortitis/genética , Aortitis/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Trasplante de Médula Ósea , Cloruro de Calcio , Línea Celular , Quimiocina CCL2/metabolismo , HDL-Colesterol/sangre , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Transducción de Señal , Factores de Tiempo , Transfección , Remodelación Vascular , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Int J Cardiol ; 243: 1-8, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28747015

RESUMEN

BACKGROUND: Biomarkers for detection of transient myocardial ischemia in patients with unstable angina (UA) or for very early diagnosis of acute myocardial infarction (AMI) are not currently available. METHODS AND RESULTS: We performed two sequential screenings of autoantibodies elevated shortly after the onset of acute coronary syndrome (ACS), and focused on metalloendopeptidase nardilysin (NRDC) among 19 identified candidate antigens. In a retrospective analysis among 93 ACS and 117 non-ACS patients, the serum level of NRDC was significantly increased in patients with ACS compared with that in patients with non-ACS (2073.5±189.8pg/ml versus 775.7±63.4pg/ml, P<0.0001). The area under the curve of NRDC for the diagnosis of ACS was 0.822 by the receiver operating characteristic curves analysis. In the time course analysis in 43 consecutive ACS patients (AMI: N=35 and UA: N=8), serum concentration of NRDC was significantly increased even in UA patients with a peak serum NRDC levels reached at admission both in AMI and UA patients. In a mouse model of AMI, we found an acute increase in serum NRDC and reduced NRDC expression in ischemic regions shortly after coronary artery ligation. NRDC expression was also reduced in infarcted regions in human autopsy samples from AMI patients. Moreover, the short treatment of primary culture of rat cardiomyocytes with H2O2 or A23187 induced NRDC secretion without cell toxicity. CONCLUSION: NRDC is a promising biomarker for the early detection of ACS, even in UA patients without elevation of necrosis markers.


Asunto(s)
Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/diagnóstico , Autoanticuerpos/sangre , Metaloendopeptidasas/sangre , Anciano , Animales , Biomarcadores/sangre , Células Cultivadas , Diagnóstico Precoz , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ratas , Estudios Retrospectivos
18.
ESC Heart Fail ; 4(2): 112-121, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28451447

RESUMEN

AIMS: Recent studies have shown that serum microRNA (miR) abundance is informative for the diagnosis or prognosis of heart failure. However, the dynamics and kinetics of miRs in acute heart failure are largely unknown. Serial measurement and analysis of serum miRs changes in individuals along their therapeutic course could reduce inter-individual variation and should detect potentially important serum miRs related to disease mechanisms. Based on this concept, we profiled serum miR signatures of blood samples that were obtained sequentially on the day of admission and on hospital Day 7. METHODS AND RESULTS: This prospective, observational study included 42 consecutive acute heart failure patients (74 ± 1 years old, 24 male). From admission to Day 7, most of the patients showed clinical improvement. In such a cohort, we detected several fluctuations of serum miRs by two distinct screening methods (quantitative PCR and high-throughput sequencing). One of these fluctuating serum miRs, miR-122-5p, decreased significantly from Day 1 to Day 7 [median arbitrary unit (1st:3rd quantile value); 4.62 [2.39:12.3] to 3.07 [1.67:5.39], P = 0.007]. This fluctuation was significantly correlated with changes in serum liver function markers (estimated coefficient and 95% confidence interval; vs change in aspartate aminotransferase 1.69, 0.890-2.484, P < 0.001 and r = 0.560, vs change in alanine aminotransferase 1.09, 0.406-1.771, P = 0.007 and r = 0.428). CONCLUSIONS: The serum miR signature of patients with acute heart failure might indicate the severity of the disease or patients' response to therapeutic intervention. Notably, serum miR-122-5p levels reflect liver damage in this condition.

19.
PLoS One ; 12(3): e0172798, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28253326

RESUMEN

BACKGROUND: Despite recent progress with drug-eluting stents, restenosis and thrombosis after endovascular intervention are still major limitations in the treatment of cardiovascular diseases. These problems are possibly caused by inappropriate inhibition of neointimal formation and retardation of re-endothelialization on the surface of the stents. miR-126 has been shown to have the potential to enhance vascular endothelial cell proliferation. METHODS AND RESULTS: We designed and constructed a 27-nt double strand RNA (dsRNA) conjugated to cholesterol, which has high membrane permeability, and formed mature miR-126 after transfection. For site-specific induction of miR-126, we utilized poly (DL-lactide-co-glycolide) nanoparticles (NPs). miR-126-dsRNA-containing NPs (miR-126 NPs) significantly reduced the protein expression of a previously identified miR-126 target, SPRED1, in human umbilical vascular endothelial cells (HUVECs), and miR-126 NPs enhanced the proliferation and migration of HUVECs. On the other hand, miR-126 NPs reduced the proliferation and migration of vascular smooth muscle cells, via the suppression of IRS-1. Finally, we developed a stent system that eluted miR-126. This delivery system exhibited significant inhibition of neointimal formation in a rabbit model of restenosis. CONCLUSIONS: miR-126 NP-conjugated stents significantly inhibited the development of neointimal hyperplasia in rabbits. The present study may indicate the possibility of a novel therapeutic option to prevent restenosis after angioplasty.


Asunto(s)
Portadores de Fármacos/química , Stents Liberadores de Fármacos , MicroARNs/química , MicroARNs/genética , Nanopartículas/química , Neointima/prevención & control , Animales , Secuencia de Bases , Movimiento Celular , Proliferación Celular , Colesterol/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ácido Láctico/química , MicroARNs/metabolismo , Músculo Liso Vascular/citología , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Bicatenario/química , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Conejos
20.
Circ Res ; 120(5): 835-847, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-27920122

RESUMEN

RATIONALE: Heart failure and atherosclerosis share the underlying mechanisms of chronic inflammation followed by fibrosis. A highly conserved microRNA (miR), miR-33, is considered as a potential therapeutic target for atherosclerosis because it regulates lipid metabolism and inflammation. However, the role of miR-33 in heart failure remains to be elucidated. OBJECTIVE: To clarify the role of miR-33 involved in heart failure. METHODS AND RESULTS: We first investigated the expression levels of miR-33a/b in human cardiac tissue samples with dilated cardiomyopathy. Increased expression of miR-33a was associated with improving hemodynamic parameters. To clarify the role of miR-33 in remodeling hearts, we investigated the responses to pressure overload by transverse aortic constriction in miR-33-deficient (knockout [KO]) mice. When mice were subjected to transverse aortic constriction, miR-33 expression levels were significantly upregulated in wild-type left ventricles. There was no difference in hypertrophic responses between wild-type and miR-33KO hearts, whereas cardiac fibrosis was ameliorated in miR-33KO hearts compared with wild-type hearts. Despite the ameliorated cardiac fibrosis, miR-33KO mice showed impaired systolic function after transverse aortic constriction. We also found that cardiac fibroblasts were mainly responsible for miR-33 expression in the heart. Deficiency of miR-33 impaired cardiac fibroblast proliferation, which was considered to be caused by altered lipid raft cholesterol content. Moreover, cardiac fibroblast-specific miR-33-deficient mice also showed decreased cardiac fibrosis induced by transverse aortic constriction as systemic miR-33KO mice. CONCLUSION: Our results demonstrate that miR-33 is involved in cardiac remodeling, and it preserves lipid raft cholesterol content in fibroblasts and maintains adaptive fibrotic responses in the remodeling heart.


Asunto(s)
Colesterol/metabolismo , Microdominios de Membrana/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Miocardio/patología , Remodelación Ventricular/fisiología , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...