Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 567(7747): 187-193, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30814737

RESUMEN

Dysregulation of lipid homeostasis is a precipitating event in the pathogenesis and progression of hepatosteatosis and metabolic syndrome. These conditions are highly prevalent in developed societies and currently have limited options for diagnostic and therapeutic intervention. Here, using a proteomic and lipidomic-wide systems genetic approach, we interrogated lipid regulatory networks in 107 genetically distinct mouse strains to reveal key insights into the control and network structure of mammalian lipid metabolism. These include the identification of plasma lipid signatures that predict pathological lipid abundance in the liver of mice and humans, defining subcellular localization and functionality of lipid-related proteins, and revealing functional protein and genetic variants that are predicted to modulate lipid abundance. Trans-omic analyses using these datasets facilitated the identification and validation of PSMD9 as a previously unknown lipid regulatory protein. Collectively, our study serves as a rich resource for probing mammalian lipid metabolism and provides opportunities for the discovery of therapeutic agents and biomarkers in the setting of hepatic lipotoxicity.


Asunto(s)
Metabolismo de los Lípidos/genética , Lípidos/análisis , Lípidos/genética , Proteómica , Animales , Células HEK293 , Humanos , Metabolismo de los Lípidos/fisiología , Lípidos/sangre , Lípidos/clasificación , Hígado/química , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Obesidad/genética , Obesidad/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo
2.
Nat Rev Mol Cell Biol ; 13(4): 213-24, 2012 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-22414897

RESUMEN

Nuclear receptors are integrators of hormonal and nutritional signals, mediating changes to metabolic pathways within the body. Given that modulation of lipid and glucose metabolism has been linked to diseases including type 2 diabetes, obesity and atherosclerosis, a greater understanding of pathways that regulate metabolism in physiology and disease is crucial. The liver X receptors (LXRs) and the farnesoid X receptors (FXRs) are activated by oxysterols and bile acids, respectively. Mounting evidence indicates that these nuclear receptors have essential roles, not only in the regulation of cholesterol and bile acid metabolism but also in the integration of sterol, fatty acid and glucose metabolism.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Receptores Nucleares Huérfanos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Aterosclerosis/metabolismo , Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Receptores X del Hígado , Obesidad/metabolismo , Receptores Nucleares Huérfanos/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Esteroles/metabolismo , Transcripción Genética
3.
Arterioscler Thromb Vasc Biol ; 41(9): 2494-2508, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34233476

RESUMEN

Objective: Low HDL-C (high-density lipoprotein cholesterol) is the most frequent dyslipidemia in Mexicans, but few studies have examined the underlying genetic basis. Our purpose was to identify genetic variants associated with HDL-C levels and cardiovascular risk in the Mexican population. Approach and Results: A genome-wide association studies for HDL-C levels in 2335 Mexicans, identified four loci associated with genome-wide significance: CETP, ABCA1, LIPC, and SIDT2. The SIDT2 missense Val636Ile variant was associated with HDL-C levels and was replicated in 3 independent cohorts (P=5.9×10−18 in the conjoint analysis). The SIDT2/Val636Ile variant is more frequent in Native American and derived populations than in other ethnic groups. This variant was also associated with increased ApoA1 and glycerophospholipid serum levels, decreased LDL-C (low-density lipoprotein cholesterol) and ApoB levels, and a lower risk of premature CAD. Because SIDT2 was previously identified as a protein involved in sterol transport, we tested whether the SIDT2/Ile636 protein affected this function using an in vitro site-directed mutagenesis approach. The SIDT2/Ile636 protein showed increased uptake of the cholesterol analog dehydroergosterol, suggesting this variant affects function. Finally, liver transcriptome data from humans and the Hybrid Mouse Diversity Panel are consistent with the involvement of SIDT2 in lipid and lipoprotein metabolism. Conclusions: This is the first genome-wide association study for HDL-C levels seeking associations with coronary artery disease in the Mexican population. Our findings provide new insight into the genetic architecture of HDL-C and highlight SIDT2 as a new player in cholesterol and lipoprotein metabolism in humans.


Asunto(s)
HDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/genética , Hiperlipoproteinemia Tipo II/genética , Proteínas de Transporte de Nucleótidos/genética , Polimorfismo de Nucleótido Simple , Adulto , Edad de Inicio , Animales , Biomarcadores/sangre , Estudios de Casos y Controles , Niño , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/epidemiología , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células HEK293 , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/epidemiología , Masculino , Análisis de la Aleatorización Mendeliana , México/epidemiología , Ratones , Persona de Mediana Edad , Proteínas de Transporte de Nucleótidos/metabolismo , Fenotipo , Medición de Riesgo
4.
Clin Sci (Lond) ; 135(24): 2691-2708, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34908104

RESUMEN

The escalating prevalence of individuals becoming overweight and obese is a rapidly rising global health problem, placing an enormous burden on health and economic systems worldwide. Whilst obesity has well described lifestyle drivers, there is also a significant and poorly understood component that is regulated by genetics. Furthermore, there is clear evidence for sexual dimorphism in obesity, where overall risk, degree, subtype and potential complications arising from obesity all differ between males and females. The molecular mechanisms that dictate these sex differences remain mostly uncharacterised. Many studies have demonstrated that this dimorphism is unable to be solely explained by changes in hormones and their nuclear receptors alone, and instead manifests from coordinated and highly regulated gene networks, both during development and throughout life. As we acquire more knowledge in this area from approaches such as large-scale genomic association studies, the more we appreciate the true complexity and heterogeneity of obesity. Nevertheless, over the past two decades, researchers have made enormous progress in this field, and some consistent and robust mechanisms continue to be established. In this review, we will discuss some of the proposed mechanisms underlying sexual dimorphism in obesity, and discuss some of the key regulators that influence this phenomenon.


Asunto(s)
Tejido Adiposo Blanco/fisiopatología , Obesidad/genética , Caracteres Sexuales , Tejido Adiposo Blanco/metabolismo , Femenino , Humanos , Masculino , Obesidad/fisiopatología
5.
Heart Lung Circ ; 29(11): 1588-1595, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32839116

RESUMEN

BACKGROUND: Cardiovascular disease is the leading cause of death in Australia. Investment in research solutions has been demonstrated to yield health and a 9.8-fold return economic benefit. The sector, however, is severely challenged with success rates of traditional peer-reviewed funding in decline. Here, we aimed to understand the perceived challenges faced by the cardiovascular workforce in Australia prior to the COVID-19 pandemic. METHODS: We used an online survey distributed across Australian cardiovascular societies/councils, universities and research institutes over a period of 6 months during 2019, with 548 completed responses. Inclusion criteria included being an Australian resident or an Australian citizen who lived overseas, and a current or past student or employee in the field of cardiovascular research. RESULTS: The mean age of respondents was 42±13 years, 47% were male, 85% had a full-time position, and 40% were a group leader or laboratory head. Twenty-three per cent (23%) had permanent employment, and 82% of full-time workers regularly worked >40 hours/week. Sixty-eight per cent (68%) said they had previously considered leaving the cardiovascular research sector. If their position could not be funded in the next few years, a staggering 91% of respondents would leave the sector. Compared to PhD- and age-matched men, women were less likely to be a laboratory head and to feel they had a long-term career path as a cardiovascular researcher, while more women were unsure about future employment and had considered leaving the sector (all p<0.05). Greater job security (76%) and government and philanthropic investment in cardiovascular research (72%) were highlighted by responders as the main changes to current practices that would encourage them to stay. CONCLUSION: Strategic solutions, such as diversification of career pathways and funding sources, and moving from a competitive to a collaborative culture, need to be a priority to decrease reliance on government funding and allow cardiovascular researchers to thrive.


Asunto(s)
Investigación Biomédica , Enfermedades Cardiovasculares , Infecciones por Coronavirus/epidemiología , Administración Financiera , Neumonía Viral/epidemiología , Investigadores , Apoyo a la Investigación como Asunto , Recursos Humanos , Adulto , Australia , Betacoronavirus , Investigación Biomédica/economía , Investigación Biomédica/organización & administración , Investigación Biomédica/tendencias , COVID-19 , Empleo/economía , Empleo/psicología , Femenino , Administración Financiera/métodos , Administración Financiera/organización & administración , Administración Financiera/estadística & datos numéricos , Financiación Gubernamental , Humanos , Masculino , Cultura Organizacional , Pandemias , Técnicas de Planificación , Investigadores/economía , Investigadores/psicología , Investigadores/estadística & datos numéricos , Apoyo a la Investigación como Asunto/organización & administración , Apoyo a la Investigación como Asunto/tendencias , SARS-CoV-2 , Encuestas y Cuestionarios , Recursos Humanos/estadística & datos numéricos
6.
Am J Physiol Endocrinol Metab ; 316(2): E293-E304, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30512991

RESUMEN

Mitochondrial dynamics refers to the constant remodeling of mitochondrial populations by multiple cellular pathways that help maintain mitochondrial health and function. Disruptions in mitochondrial dynamics often lead to mitochondrial dysfunction, which is frequently associated with disease in rodents and humans. Consistent with this, obesity is associated with reduced mitochondrial function in white adipose tissue, partly via alterations in mitochondrial dynamics. Several proteins, including the E3 ubiquitin ligase membrane-associated RING-CH-type finger 5 (MARCH5), are known to regulate mitochondrial dynamics; however, the role of these proteins in adipocytes has been poorly studied. Here, we show that MARCH5 is regulated by peroxisome proliferator-activated receptor-γ (PPARγ) during adipogenesis and is correlated with fat mass across a panel of genetically diverse mouse strains, in ob/ob mice, and in humans. Furthermore, manipulation of MARCH5 expression in vitro and in vivo alters mitochondrial function, affects cellular metabolism, and leads to differential regulation of several metabolic genes. Thus our data demonstrate an association between mitochondrial dynamics and metabolism that defines MARCH5 as a critical link between these interconnected pathways.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Proteínas de la Membrana/metabolismo , Síndrome Metabólico/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células 3T3-L1 , Adipogénesis , Adulto , Animales , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Mitocondriales/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Ubiquitina-Proteína Ligasas/genética
7.
Genes Dev ; 25(12): 1262-74, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21685362

RESUMEN

We previously identified the E3 ubiquitin ligase IDOL as a sterol-dependent regulator of the LDL receptor (LDLR). The molecular pathway underlying IDOL action, however, remains to be determined. Here we report the identification and biochemical and structural characterization of an E2-E3 ubiquitin ligase complex for LDLR degradation. We identified the UBE2D family (UBE2D1-4) as E2 partners for IDOL that support both autoubiquitination and IDOL-dependent ubiquitination of the LDLR in a cell-free system. NMR chemical shift mapping and a 2.1 Å crystal structure of the IDOL RING domain-UBE2D1 complex revealed key interactions between the dimeric IDOL protein and the E2 enzyme. Analysis of the IDOL-UBE2D1 interface also defined the stereochemical basis for the selectivity of IDOL for UBE2Ds over other E2 ligases. Structure-based mutations that inhibit IDOL dimerization or IDOL-UBE2D interaction block IDOL-dependent LDLR ubiquitination and degradation. Furthermore, expression of a dominant-negative UBE2D enzyme inhibits the ability of IDOL to degrade the LDLR in cells. These results identify the IDOL-UBE2D complex as an important determinant of LDLR activity, and provide insight into molecular mechanisms underlying the regulation of cholesterol uptake.


Asunto(s)
Receptores de LDL/metabolismo , Esteroles/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Células HEK293 , Humanos , Hierro/metabolismo , Modelos Moleculares , Estructura Terciaria de Proteína , Estereoisomerismo , Especificidad por Sustrato , Enzimas Ubiquitina-Conjugadoras/química , Ubiquitina-Proteína Ligasas/química
8.
Clin Sci (Lond) ; 131(12): 1245-1260, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28592700

RESUMEN

Cardiovascular events are the major cause of morbidity and mortality in Type 2 diabetes (T2D). This condition is associated with heightened platelet reactivity, contributing to increased atherothrombotic risk. Indeed, individuals with diabetes respond inadequately to standard antiplatelet therapy. Furthermore, they often experience recurrent events as well as side effects that include excess bleeding. This highlights the need for identification of novel regulators of diabetes-associated thrombosis to target for therapeutic intervention. It is well established that platelet aggregation, a process essential for thrombus formation, is tightly regulated by shear stress; however, the mechanisms underlying shear activation of platelets, particularly in the setting of diabetes, are still poorly understood. This review will address the limitations of current diagnostic systems to assess the importance of shear stress in the regulation of thrombus formation in T2D, and the inability to recapitulate the pro-thrombotic phenotype seen clinically in the setting of T2D. Moreover, we will discuss recent findings utilizing new technologies to define the importance of shear stress in thrombus formation and their potential application to the setting of diabetes. Finally, we will discuss the potential of targeting shear-dependent mechanisms of thrombus formation as a novel therapeutic approach in the setting of T2D.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Mecanotransducción Celular , Activación Plaquetaria , Trombosis/etiología , Animales , Glucemia/metabolismo , Plaquetas/efectos de los fármacos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/mortalidad , Resistencia a Medicamentos , Células Endoteliales/metabolismo , Fibrinolíticos/uso terapéutico , Humanos , Mecanotransducción Celular/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/uso terapéutico , Flujo Sanguíneo Regional , Estrés Mecánico , Trombosis/sangre , Trombosis/mortalidad , Trombosis/prevención & control , Factor de von Willebrand/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 36(2): 295-307, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26715682

RESUMEN

OBJECTIVE: Oxidative stress is considered a hallmark of atherosclerosis. In particular, the superoxide-generating type 1 NADPH oxidase (NOX1) has been shown to be induced and play a pivotal role in early phases of mouse models of atherosclerosis and in the context of diabetes mellitus. Here, we investigated the role of the most abundant type 4 isoform (NOX4) in human and mouse advanced atherosclerosis. APPROACH AND RESULTS: Plaques of patients with cardiovascular events or established diabetes mellitus showed a surprising reduction in expression of the most abundant but hydrogen peroxide (H2O2)-generating type 4 isoform (Nox4), whereas Nox1 mRNA was elevated, when compared with respective controls. As these data suggested that NOX4-derived reactive oxygen species may convey a surprisingly protective effect during plaque progression, we examined a mouse model of accelerated and advanced diabetic atherosclerosis, the streptozotocin-treated ApoE(-/-) mouse, with (NOX4(-/-)) and without genetic deletion of Nox4. Similar to the human data, advanced versus early plaques of wild-type mice showed reduced Nox4 mRNA expression. Consistent with a rather protective role of NOX4-derived reactive oxygen species, NOX4(-/-) mice showed increased atherosclerosis when compared with wild-type mice. Deleting NOX4 was associated with reduced H2O2 forming activity and attenuation of the proinflammatory markers, monocyte chemotratic protein-1, interleukin-1ß, and tumor necrosis factor-α, as well as vascular macrophage accumulation. Furthermore, there was a greater accumulation of fibrillar collagen fibres within the vascular wall and plaque in diabetic Nox4(-/-)ApoE(-/-) mice, indicative of plaque remodeling. These data could be replicated in human aortic endothelial cells in vitro, where Nox4 overexpression increased H2O2 and reduced the expression of pro-oxidants and profibrotic markers. Interestingly, Nox4 levels inversely correlated with Nox2 gene and protein levels. Although NOX2 is not constitutively active unlike NOX4 and forms rather superoxide, this opens up the possibility that at least some effects of NOX4 deletion are mediated by NOX2 activation. CONCLUSIONS: Thus, the appearance of reactive oxygen species in atherosclerosis is apparently not always a nondesirable oxidative stress, but can also have protective effects. Both in humans and in mouse, the H2O2-forming NOX4, unlike the superoxide-forming NOX1, can act as a negative modulator of inflammation and remodeling and convey atheroprotection. These results have implications on how to judge reactive oxygen species formation in cardiovascular disease and need to be considered in the development of NOX inhibitory drugs.


Asunto(s)
Aorta/enzimología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Peróxido de Hidrógeno/metabolismo , Inflamación/prevención & control , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Placa Aterosclerótica , Remodelación Vascular , Animales , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Estudios de Casos y Controles , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Colágenos Fibrilares/metabolismo , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Noqueados , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , Interferencia de ARN , Transducción de Señal , Superóxidos/metabolismo , Factores de Tiempo , Transfección
10.
J Biol Chem ; 290(9): 5566-81, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25468909

RESUMEN

Obesity is associated with increased breast cancer (BrCA) incidence. Considering that inactivation of estrogen receptor (ER)α promotes obesity and metabolic dysfunction in women and female mice, understanding the mechanisms and tissue-specific sites of ERα action to combat metabolic-related disease, including BrCA, is of clinical importance. To study the role of ERα in adipose tissue we generated fat-specific ERα knock-out (FERKO) mice. Herein we show that ERα deletion increased adipocyte size, fat pad weight, and tissue expression and circulating levels of the secreted glycoprotein, lipocalin 2 (Lcn2), an adipokine previously associated with BrCA development. Chromatin immunoprecipitation and luciferase reporter studies showed that ERα binds the Lcn2 promoter to repress its expression. Because adipocytes constitute an important cell type of the breast microenvironment, we examined the impact of adipocyte ERα deletion on cancer cell behavior. Conditioned medium from ERα-null adipocytes and medium containing pure Lcn2 increased proliferation and migration of a subset of BrCA cells in culture. The proliferative and promigratory effects of ERα-deficient adipocyte-conditioned medium on BrCA cells was reversed by Lcn2 deletion. BrCA cell responsiveness to exogenous Lcn2 was heightened in cell types where endogenous Lcn2 expression was minimal, but components of the Lcn2 signaling pathway were enriched, i.e. SLC22A17 and 3-hydroxybutyrate dehydrogenase (BDH2). In breast tumor biopsies from women diagnosed with BrCA we found that BDH2 expression was positively associated with adiposity and circulating Lcn2 levels. Collectively these data suggest that reduction of ERα expression in adipose tissue promotes adiposity and is linked with the progression and severity of BrCA via increased adipocyte-specific Lcn2 production and enhanced tumor cell Lcn2 sensitivity.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Tejido Adiposo/metabolismo , Receptor alfa de Estrógeno/metabolismo , Lipocalinas/metabolismo , Obesidad/metabolismo , Proteínas Oncogénicas/metabolismo , Células 3T3-L1 , Proteínas de Fase Aguda/genética , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Immunoblotting , Lipocalina 2 , Lipocalinas/sangre , Lipocalinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Proteínas Oncogénicas/sangre , Proteínas Oncogénicas/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Circ Res ; 115(4): 442-9, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24935961

RESUMEN

RATIONALE: The E3 ubiquitin ligase inducible degrader of the low-density lipoprotein receptor (IDOL) triggers lysosomal degradation of the low-density lipoprotein receptor. The tissue-specific effects of the IDOL pathway on plasma cholesterol and atherosclerosis have not been examined. OBJECTIVE: Given that the liver is the primary determinant of plasma cholesterol levels, we sought to examine the consequence of effect of chronic liver-specific expression of a dominant-active form of IDOL in mice. METHODS AND RESULTS: We expressed a degradation-resistant, dominant-active form of IDOL (super IDOL [sIDOL]) in C57Bl/6J mice from the liver-specific albumin promoter (L-sIDOL transgenics). L-sIDOL mice were fed a Western diet for 20 or 30 weeks and then analyzed for plasma lipid levels and atherosclerotic lesion formation. L-sIDOL mice showed dramatic reductions in hepatic low-density lipoprotein receptor protein and increased plasma low-density lipoprotein cholesterol levels on both chow and Western diets. Moreover, L-sIDOL mice developed marked atherosclerotic lesions when fed a Western diet. Lesion formation in L-sIDOL mice was more robust than in apolipoprotein E*3 Leiden mice and did not require the addition of cholate to the diet. Western diet-fed L-sIDOL mice had elevated expression of liver X receptor target genes and proinflammatory genes in their aortas. CONCLUSIONS: Liver-specific expression of dominant-active IDOL is associated with hypercholesterolemia and a marked elevation in atherosclerotic lesions. Our results show that increased activity of the IDOL pathway in the liver can override other low-density lipoprotein receptor regulatory pathways leading to cardiovascular disease. L-sIDOL mice are a robust, dominantly inherited, diet-inducible model for the study of atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/patología , Aterosclerosis/enzimología , Dieta Alta en Grasa , Dieta Occidental , Hipercolesterolemia/enzimología , Hígado/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Albúminas/genética , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , LDL-Colesterol/sangre , Modelos Animales de Enfermedad , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Mediadores de Inflamación/metabolismo , Receptores X del Hígado , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Receptores Nucleares Huérfanos/metabolismo , Placa Aterosclerótica , Regiones Promotoras Genéticas , Receptores de LDL/metabolismo , Factores de Tiempo , Ubiquitina-Proteína Ligasas/genética
13.
Proc Natl Acad Sci U S A ; 108(50): 20107-12, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22109552

RESUMEN

The E3 ubiquitin ligase IDOL (inducible degrader of the LDL receptor) regulates LDL receptor (LDLR)-dependent cholesterol uptake, but its mechanism of action, including the molecular basis for its stringent specificity, is poorly understood. Here we show that IDOL uses a singular strategy among E3 ligases for target recognition. The IDOL FERM domain binds directly to a recognition sequence in the cytoplasmic tails of lipoprotein receptors. This physical interaction is independent of IDOL's really interesting new gene (RING) domain E3 ligase activity and its capacity for autoubiquitination. Furthermore, IDOL controls its own stability through autoubiquitination of a unique FERM subdomain fold not present in other FERM proteins. Key residues defining the IDOL-LDLR interaction and IDOL autoubiquitination are functionally conserved in their insect homologs. Finally, we demonstrate that target recognition by IDOL involves a tripartite interaction between the FERM domain, membrane phospholipids, and the lipoprotein receptor tail. Our data identify the IDOL-LDLR interaction as an evolutionarily conserved mechanism for the regulation of lipid uptake and suggest that this interaction could potentially be exploited for the pharmacologic modulation of lipid metabolism.


Asunto(s)
Proteolisis , Receptores de Lipoproteína/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Aminoácidos , Animales , Membrana Celular/metabolismo , Secuencia Conservada , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Receptores de Lipoproteína/química , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
14.
Diabetes ; 73(9): 1495-1512, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38905153

RESUMEN

Despite advances in treatment, atherosclerotic cardiovascular disease remains the leading cause of death in patients with diabetes. Even when risk factors are mitigated, the disease progresses, and thus, newer targets need to be identified that directly inhibit the underlying pathobiology of atherosclerosis in diabetes. A single-cell sequencing approach was used to distinguish the proatherogenic transcriptional profile in aortic cells in diabetes using a streptozotocin-induced diabetic Apoe-/- mouse model. Human carotid endarterectomy specimens from individuals with and without diabetes were also evaluated via immunohistochemical analysis. Further mechanistic studies were performed in human aortic endothelial cells (HAECs) and human THP-1-derived macrophages. We then performed a preclinical study using an activator protein-1 (AP-1) inhibitor in a diabetic Apoe-/- mouse model. Single-cell RNA sequencing analysis identified the AP-1 complex as a novel target in diabetes-associated atherosclerosis. AP-1 levels were elevated in carotid endarterectomy specimens from individuals with diabetes compared with those without diabetes. AP-1 was validated as a mechanosensitive transcription factor via immunofluorescence staining for regional heterogeneity of endothelial cells of the aortic region exposed to turbulent blood flow and by performing microfluidics experiments in HAECs. AP-1 inhibition with T-5224 blunted endothelial cell activation as assessed by a monocyte adhesion assay and expression of genes relevant to endothelial function. Furthermore, AP-1 inhibition attenuated foam cell formation. Critically, treatment with T-5224 attenuated atherosclerosis development in diabetic Apoe-/- mice. This study has identified the AP-1 complex as a novel target, the inhibition of which treats the underlying pathobiology of atherosclerosis in diabetes.


Asunto(s)
Aterosclerosis , Diabetes Mellitus Experimental , Análisis de la Célula Individual , Factor de Transcripción AP-1 , Animales , Aterosclerosis/metabolismo , Aterosclerosis/genética , Humanos , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/genética , Ratones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/complicaciones , Masculino , Células Endoteliales/metabolismo , Análisis de Secuencia de ARN
15.
J Endocrinol ; 259(2)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37725394

RESUMEN

Type 2 diabetes mellitus (T2DM), a condition characterised by insulin resistance (IR) and skeletal muscle mitochondrial abnormalities, is a leading cause of death in developed societies. Much work has postulated that improving pathways linked to mitochondrial health, including autophagy, may be a potential avenue to prevent or treat T2DM. Given the recent data indicating a role for tripartite motif-containing 28 (TRIM28) in autophagy and mitochondrial pathways, we investigated whether muscle-specific deletion of TRIM28 might impact on obesity, glucose tolerance, and IR in mice. We studied two different muscle-specific (MCK-cre and ACTA1-cre-ERT2) TRIM28 knockout models, which were phenotyped during and after being fed a chow or high-fat diet (HFD). Whilst muscle-specific deletion of TRIM28 in both models demonstrated alterations in markers of mitochondrial activity and autophagy in skeletal muscle, we did not observe major impacts on the majority of metabolic measures in these mice. Specifically, we demonstrate that deletion of TRIM28 in skeletal muscle of mice during (MCK-cre) or post-development (ACTA1-cre-ERT2) does not prevent HFD-induced obesity or glucose intolerance. These findings are consistent with those reported previously in relation to autophagy and mitochondria in other cell types, and thus warrant further study into the biological role TRIM28 has in relation to mitochondrial function.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Resistencia a la Insulina , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/genética , Músculo Esquelético/metabolismo , Intolerancia a la Glucosa/metabolismo , Obesidad/metabolismo , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Mitocondrias Musculares/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo
16.
Elife ; 122023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-37000167

RESUMEN

Cardiometabolic diseases encompass a range of interrelated conditions that arise from underlying metabolic perturbations precipitated by genetic, environmental, and lifestyle factors. While obesity, dyslipidaemia, smoking, and insulin resistance are major risk factors for cardiometabolic diseases, individuals still present in the absence of such traditional risk factors, making it difficult to determine those at greatest risk of disease. Thus, it is crucial to elucidate the genetic, environmental, and molecular underpinnings to better understand, diagnose, and treat cardiometabolic diseases. Much of this information can be garnered using systems genetics, which takes population-based approaches to investigate how genetic variance contributes to complex traits. Despite the important advances made by human genome-wide association studies (GWAS) in this space, corroboration of these findings has been hampered by limitations including the inability to control environmental influence, limited access to pertinent metabolic tissues, and often, poor classification of diseases or phenotypes. A complementary approach to human GWAS is the utilisation of model systems such as genetically diverse mouse panels to study natural genetic and phenotypic variation in a controlled environment. Here, we review mouse genetic reference panels and the opportunities they provide for the study of cardiometabolic diseases and related traits. We discuss how the post-GWAS era has prompted a shift in focus from discovery of novel genetic variants to understanding gene function. Finally, we highlight key advantages and challenges of integrating complementary genetic and multi-omics data from human and mouse populations to advance biological discovery.


Asunto(s)
Enfermedades Cardiovasculares , Estudio de Asociación del Genoma Completo , Animales , Humanos , Ratones , Enfermedades Cardiovasculares/genética , Predisposición Genética a la Enfermedad , Obesidad/genética , Fenotipo , Factores de Riesgo
17.
Physiol Rep ; 11(23): e15869, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38054572

RESUMEN

Long ncRNAs (lncRNAs) have been shown to play a biological and physiological role in various tissues including the heart. We and others have previously established that the lncRNA Oip5os1 (1700020I14Rik, OIP5-AS1, Cyrano) is enriched in striated muscles, and its deletion in mice leads to defects in both skeletal and cardiac muscle function. In the present study, we investigated the impact of global Oip5os1 deletion on cardiac function in the setting of streptozotocin (STZ)-induced diabetes. Specifically, we studied male WT and KO mice with or without diabetes for 24 weeks, and phenotyped animals for metabolic and cardiac endpoints. Independent of genotype, diabetes was associated with left ventricular diastolic dysfunction based on a fall in E'/A' ratio. Deletion of Oip5os1 in a setting of diabetes had no significant impact on ventricular function or ventricular weight, but was associated with left atrial dysfunction (reduced fractional shortening) and myopathy which was associated with anesthesia intolerance and premature death in the majority of KO mice tested during cardiac functional assessment. This atrial phenotype was not observed in WT diabetic mice. The most striking molecular difference was a reduction in the metabolic regulator ERRalpha in the atria of KO mice compared with WT mice. There was also a trend for a reduction in Serca2a. These findings highlight Oip5os1 as a gene of interest in aspects of atrial function in the setting of diabetes, highlighting an additional functional role for this lncRNA in cardiac pathological settings.


Asunto(s)
Fibrilación Atrial , Diabetes Mellitus Experimental , ARN Largo no Codificante , Animales , Masculino , Ratones , Fibrilación Atrial/complicaciones , Fibrilación Atrial/genética , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Miocardio/patología , ARN Largo no Codificante/genética
18.
Front Physiol ; 13: 837001, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35283789

RESUMEN

Adipose tissue is comprised of a heterogeneous population of cells that co-operate to perform diverse physiological roles including endocrine-related functions. The endocrine role of adipose tissue enables it to communicate nutritional and health cues to other organs, such as the liver, muscle, and brain, in order to regulate appetite and whole body metabolism. Adipose tissue dysfunction, which is often observed in obesity, is associated with changes in the adipose secretome, which can subsequently contribute to disease pathology. Indeed, secreted bioactive factors released from adipose tissue contribute to metabolic homeostasis and likely play a causal role in disease; however, what constitutes the entirety of the adipose tissue secretome is still poorly understood. Recent advances in nanotechnology have advanced this field substantially and have led to the identification of small, secreted particles known as extracellular vesicles (EVs). These small nano-sized lipid envelopes are released by most cell types and are capable of systemically delivering bioactive molecules, such as nucleic acids, proteins, and lipids. EVs interact with target cells to deliver specific cargo that can then elicit effects in various tissues throughout the body. Adipose tissue has recently been shown to secrete EVs that can communicate with the periphery to maintain metabolic homeostasis, or under certain pathological conditions, drive disease. In this review, we discuss the current landscape of adipose tissue-derived EVs, with a focus on their role in the regulation of metabolic homeostasis and disease pathology.

19.
Front Physiol ; 13: 839471, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35295579

RESUMEN

Synthetic oligonucleotide technologies are DNA or RNA based molecular compounds that are utilized to disrupt gene transcription or translation in target tissues or cells. Optimally, oligonucleotides are 10-30 base pairs in length, and mediate target gene suppression through directed sequence homology with messenger RNA (mRNA), leading to mRNA degradation. Examples of specific oligonucleotide technologies include antisense oligonucleotides (ASO), short hairpin RNAs (shRNA), and small interfering RNAs (siRNA). In vitro and in vivo studies that model obesity related disorders have demonstrated that oligonucleotide technologies can be implemented to improve the metabolism of cells and tissues, exemplified by improvements in fat utilization and hepatic insulin signaling, respectively. Oligonucleotide therapy has also been associated with reductions in lipid accumulation in both the liver and adipose tissue in models of diet-induced obesity. Recent advances in oligonucleotide technologies include the addition of chemical modifications such as N-acetylgalactosamine (GalNAc) conjugates that have been successful at achieving affinity for the liver, in turn improving specificity, and thus reducing off target effects. However, some challenges are still yet to be overcome relating to hepatic injury and off-target effects that have been reported with some compounds, including ASOs. In summary, oligonucleotide-based therapies are an effective tool to elucidate mechanistic insights into metabolic pathways and provide an attractive avenue for translational research into the clinic.

20.
Front Physiol ; 13: 841278, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309067

RESUMEN

Ether lipids are a unique class of glycero- and glycerophospho-lipid that carry an ether or vinyl ether linked fatty alcohol at the sn-1 position of the glycerol backbone. These specialised lipids are important endogenous anti-oxidants with additional roles in regulating membrane fluidity and dynamics, intracellular signalling, immunomodulation and cholesterol metabolism. Lipidomic profiling of human population cohorts has identified new associations between reduced circulatory plasmalogen levels, an abundant and biologically active sub-class of ether lipids, with obesity and body-mass index. These findings align with the growing body of work exploring novel roles for ether lipids within adipose tissue. In this regard, ether lipids have now been linked to facilitating lipid droplet formation, regulating thermogenesis and mediating beiging of white adipose tissue in early life. This review will assess recent findings in both population studies and studies using cell and animal models to delineate the functional and protective roles of ether lipids in the setting of obesity. We will also discuss the therapeutic potential of ether lipid supplementation to attenuate diet-induced obesity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA