Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(16)2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37629051

RESUMEN

Obesity is a growing public health problem associated with increased risk of type 2 diabetes, cardiovascular disease, nonalcoholic fatty liver disease (NAFLD) and cancer. Here, we identify microRNA-22 (miR-22) as an essential rheostat involved in the control of lipid and energy homeostasis as well as the onset and maintenance of obesity. We demonstrate through knockout and transgenic mouse models that miR-22 loss-of-function protects against obesity and hepatic steatosis, while its overexpression promotes both phenotypes even when mice are fed a regular chow diet. Mechanistically, we show that miR-22 controls multiple pathways related to lipid biogenesis and differentiation. Importantly, genetic ablation of miR-22 favors metabolic rewiring towards higher energy expenditure and browning of white adipose tissue, suggesting that modulation of miR-22 could represent a viable therapeutic strategy for treatment of obesity and other metabolic disorders.


Asunto(s)
Diabetes Mellitus Tipo 2 , MicroARNs , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Homeostasis , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , MicroARNs/genética , Lípidos
2.
Nat Commun ; 13(1): 7630, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494335

RESUMEN

Severe COVID-19 is associated with epithelial and endothelial barrier dysfunction within the lung as well as in distal organs. While it is appreciated that an exaggerated inflammatory response is associated with barrier dysfunction, the triggers of vascular leak are unclear. Here, we report that cell-intrinsic interactions between the Spike (S) glycoprotein of SARS-CoV-2 and epithelial/endothelial cells are sufficient to induce barrier dysfunction in vitro and vascular leak in vivo, independently of viral replication and the ACE2 receptor. We identify an S-triggered transcriptional response associated with extracellular matrix reorganization and TGF-ß signaling. Using genetic knockouts and specific inhibitors, we demonstrate that glycosaminoglycans, integrins, and the TGF-ß signaling axis are required for S-mediated barrier dysfunction. Notably, we show that SARS-CoV-2 infection caused leak in vivo, which was reduced by inhibiting integrins. Our findings offer mechanistic insight into SARS-CoV-2-triggered vascular leak, providing a starting point for development of therapies targeting COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Enzima Convertidora de Angiotensina 2 , Glicoproteína de la Espiga del Coronavirus/genética , Células Endoteliales , Integrinas , Peptidil-Dipeptidasa A/genética , Factor de Crecimiento Transformador beta
3.
Sci Rep ; 12(1): 18506, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36323770

RESUMEN

SARS coronavirus 2 (SARS-CoV-2) has caused an ongoing global pandemic with significant mortality and morbidity. At this time, the only FDA-approved therapeutic for COVID-19 is remdesivir, a broad-spectrum antiviral nucleoside analog. Efficacy is only moderate, and improved treatment strategies are urgently needed. To accomplish this goal, we devised a strategy to identify compounds that act synergistically with remdesivir in preventing SARS-CoV-2 replication. We conducted combinatorial high-throughput screening in the presence of submaximal remdesivir concentrations, using a human lung epithelial cell line infected with a clinical isolate of SARS-CoV-2. This identified 20 approved drugs that act synergistically with remdesivir, many with favorable pharmacokinetic and safety profiles. Strongest effects were observed with established antivirals, Hepatitis C virus nonstructural protein 5A (HCV NS5A) inhibitors velpatasvir and elbasvir. Combination with their partner drugs sofosbuvir and grazoprevir further increased efficacy, increasing remdesivir's apparent potency > 25-fold. We report that HCV NS5A inhibitors act on the SARS-CoV-2 exonuclease proofreader, providing a possible explanation for the synergy observed with nucleoside analog remdesivir. FDA-approved Hepatitis C therapeutics Epclusa® (velpatasvir/sofosbuvir) and Zepatier® (elbasvir/grazoprevir) could be further optimized to achieve potency and pharmacokinetic properties that support clinical evaluation in combination with remdesivir.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Hepatitis C , Humanos , SARS-CoV-2 , Antivirales/uso terapéutico , Sofosbuvir/farmacología , Nucleósidos/farmacología , Adenosina Monofosfato , Alanina , Hepacivirus , Hepatitis C/tratamiento farmacológico , Pulmón
4.
Nat Commun ; 13(1): 4503, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35922434

RESUMEN

The COVID-19 pandemic is exacting an increasing toll worldwide, with new SARS-CoV-2 variants emerging that exhibit higher infectivity rates and that may partially evade vaccine and antibody immunity. Rapid deployment of non-invasive therapeutic avenues capable of preventing infection by all SARS-CoV-2 variants could complement current vaccination efforts and help turn the tide on the COVID-19 pandemic. Here, we describe a novel therapeutic strategy targeting the SARS-CoV-2 RNA using locked nucleic acid antisense oligonucleotides (LNA ASOs). We identify an LNA ASO binding to the 5' leader sequence of SARS-CoV-2 that disrupts a highly conserved stem-loop structure with nanomolar efficacy in preventing viral replication in human cells. Daily intranasal administration of this LNA ASO in the COVID-19 mouse model potently suppresses viral replication (>80-fold) in the lungs of infected mice. We find that the LNA ASO is efficacious in countering all SARS-CoV-2 "variants of concern" tested both in vitro and in vivo. Hence, inhaled LNA ASOs targeting SARS-CoV-2 represents a promising therapeutic approach to reduce or prevent transmission and decrease severity of COVID-19 in infected individuals. LNA ASOs are chemically stable and can be flexibly modified to target different viral RNA sequences and could be stockpiled for future coronavirus pandemics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Administración Intranasal , Animales , Humanos , Ratones , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Pandemias/prevención & control , ARN Viral/genética
5.
Mol Ther ; 29(7): 2281-2293, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-33744470

RESUMEN

Abnormal cholesterol/lipid homeostasis is linked to neurodegenerative conditions such as age-related macular degeneration (AMD), which is a leading cause of blindness in the elderly. The most prevalent form, termed "dry" AMD, is characterized by pathological cholesterol accumulation beneath the retinal pigment epithelial (RPE) cell layer and inflammation-linked degeneration in the retina. We show here that the cholesterol-regulating microRNA miR-33 was elevated in the RPE of aging mice. Expression of the miR-33 target ATP-binding cassette transporter (ABCA1), a cholesterol efflux pump genetically linked to AMD, declined reciprocally in the RPE with age. In accord, miR-33 modulated ABCA1 expression and cholesterol efflux in human RPE cells. Subcutaneous delivery of miR-33 antisense oligonucleotides (ASO) to aging mice and non-human primates fed a Western-type high fat/cholesterol diet resulted in increased ABCA1 expression, decreased cholesterol accumulation, and reduced immune cell infiltration in the RPE cell layer, accompanied by decreased pathological changes to RPE morphology. These findings suggest that miR-33 targeting may decrease cholesterol deposition and ameliorate AMD initiation and progression.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Colesterol/metabolismo , Inflamación/terapia , Degeneración Macular/terapia , MicroARNs/antagonistas & inhibidores , Fenotipo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Inflamación/etiología , Inflamación/patología , Macaca fascicularis , Degeneración Macular/etiología , Degeneración Macular/patología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Oligonucleótidos Antisentido/genética
6.
iScience ; 24(2): 102021, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33426509

RESUMEN

The unparalleled global effort to combat the continuing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic over the last year has resulted in promising prophylactic measures. However, a need still exists for cheap, effective therapeutics, and targeting multiple points in the viral life cycle could help tackle the current, as well as future, coronaviruses. Here, we leverage our recently developed, ultra-large-scale in silico screening platform, VirtualFlow, to search for inhibitors that target SARS-CoV-2. In this unprecedented structure-based virtual campaign, we screened roughly 1 billion molecules against each of 40 different target sites on 17 different potential viral and host targets. In addition to targeting the active sites of viral enzymes, we also targeted critical auxiliary sites such as functionally important protein-protein interactions.

7.
Cancer Discov ; 11(3): 678-695, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33203734

RESUMEN

Circulating tumor cells (CTC) are shed by cancer into the bloodstream, where a viable subset overcomes oxidative stress to initiate metastasis. We show that single CTCs from patients with melanoma coordinately upregulate lipogenesis and iron homeostasis pathways. These are correlated with both intrinsic and acquired resistance to BRAF inhibitors across clonal cultures of BRAF-mutant CTCs. The lipogenesis regulator SREBP2 directly induces transcription of the iron carrier Transferrin (TF), reducing intracellular iron pools, reactive oxygen species, and lipid peroxidation, thereby conferring resistance to inducers of ferroptosis. Knockdown of endogenous TF impairs tumor formation by melanoma CTCs, and their tumorigenic defects are partially rescued by the lipophilic antioxidants ferrostatin-1 and vitamin E. In a prospective melanoma cohort, presence of CTCs with high lipogenic and iron metabolic RNA signatures is correlated with adverse clinical outcome, irrespective of treatment regimen. Thus, SREBP2-driven iron homeostatic pathways contribute to cancer progression, drug resistance, and metastasis. SIGNIFICANCE: Through single-cell analysis of primary and cultured melanoma CTCs, we have uncovered intrinsic cancer cell heterogeneity within lipogenic and iron homeostatic pathways that modulates resistance to BRAF inhibitors and to ferroptosis inducers. Activation of these pathways within CTCs is correlated with adverse clinical outcome, pointing to therapeutic opportunities.This article is highlighted in the In This Issue feature, p. 521.


Asunto(s)
Ferroptosis/genética , Lipogénesis/genética , Melanoma/genética , Melanoma/metabolismo , Células Neoplásicas Circulantes/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Transferrina/metabolismo , Biomarcadores de Tumor , Células Cultivadas , Susceptibilidad a Enfermedades , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Melanoma/patología , Mutación , Células Neoplásicas Circulantes/patología , Transducción de Señal , Análisis de la Célula Individual , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
8.
ChemRxiv ; 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-33200116

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), previously known as 2019 novel coronavirus (2019-nCoV), has spread rapidly across the globe, creating an unparalleled global health burden and spurring a deepening economic crisis. As of July 7th, 2020, almost seven months into the outbreak, there are no approved vaccines and few treatments available. Developing drugs that target multiple points in the viral life cycle could serve as a strategy to tackle the current as well as future coronavirus pandemics. Here we leverage the power of our recently developed in silico screening platform, VirtualFlow, to identify inhibitors that target SARS-CoV-2. VirtualFlow is able to efficiently harness the power of computing clusters and cloud-based computing platforms to carry out ultra-large scale virtual screens. In this unprecedented structure-based multi-target virtual screening campaign, we have used VirtualFlow to screen an average of approximately 1 billion molecules against each of 40 different target sites on 17 different potential viral and host targets in the cloud. In addition to targeting the active sites of viral enzymes, we also target critical auxiliary sites such as functionally important protein-protein interaction interfaces. This multi-target approach not only increases the likelihood of finding a potent inhibitor, but could also help identify a collection of anti-coronavirus drugs that would retain efficacy in the face of viral mutation. Drugs belonging to different regimen classes could be combined to develop possible combination therapies, and top hits that bind at highly conserved sites would be potential candidates for further development as coronavirus drugs. Here, we present the top 200 in silico hits for each target site. While in-house experimental validation of some of these compounds is currently underway, we want to make this array of potential inhibitor candidates available to researchers worldwide in consideration of the pressing need for fast-tracked drug development.

9.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33058756

RESUMEN

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Asunto(s)
Enfermedades Metabólicas/genética , MicroARNs/genética , Adipocitos Marrones/patología , Adiposidad , Alelos , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Dieta Alta en Grasa , Metabolismo Energético , Epigénesis Genética , Sitios Genéticos , Glucosa/metabolismo , Homeostasis , Humanos , Hipertrofia , Resistencia a la Insulina , Leptina/deficiencia , Leptina/metabolismo , Masculino , Mamíferos/genética , Ratones Endogámicos C57BL , Ratones Obesos , MicroARNs/metabolismo , Obesidad/genética , Oligonucleótidos/metabolismo , Especificidad de la Especie
10.
Sci Rep ; 9(1): 10369, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316083

RESUMEN

de novo fatty acid biosynthesis (DNFA) is a hallmark adaptation of many cancers that supports survival, proliferation, and metastasis. Here we elucidate previously unexplored aspects of transcription regulation and clinical relevance of DNFA in cancers. We show that elevated expression of DNFA genes is characteristic of many tumor types and correlates with poor prognosis, especially in melanomas. Elevated DNFA gene expression depends on the SREBP1 transcription factor in multiple melanoma cell lines. SREBP1 predominantly binds to the transcription start sites of DNFA genes, regulating their expression by recruiting RNA polymerase II to promoters for productive transcription elongation. We find that SREBP1-regulated DNFA represents a survival trait in melanoma cells, regardless of proliferative state and oncogenic mutation status. Indeed, malignant melanoma cells exhibit elevated DNFA gene expression after the BRAF/MEK signaling pathway is blocked (e.g. by BRAF inhibitors), and DNFA expression remains higher in melanoma cells resistant to vemurafenib treatment than in untreated cells. Accordingly, DNFA pathway inhibition, whether by direct targeting of SREBP1 with antisense oligonucleotides, or through combinatorial effects of multiple DNFA enzyme inhibitors, exerts potent cytotoxic effects on both BRAFi-sensitive and -resistant melanoma cells. Altogether, these results implicate SREBP1 and DNFA enzymes as enticing therapeutic targets in melanomas.


Asunto(s)
Ácidos Grasos/biosíntesis , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas de Neoplasias/fisiología , Neoplasias Cutáneas/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Línea Celular Tumoral , Supervivencia Celular , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indazoles/farmacología , Estimación de Kaplan-Meier , Melanoma/genética , Melanoma/mortalidad , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/genética , Piperazinas/farmacología , Pronóstico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Transcripción Genética , Vemurafenib/farmacología
11.
PLoS One ; 14(4): e0215022, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30970006

RESUMEN

While investigating the role played by de novo lipid (DNL) biosynthesis in cancer cells, we sought a medium condition that would support cell proliferation without providing any serum lipids. Here we report that a defined serum free cell culture medium condition containing insulin, transferrin and selenium (ITS) supports controlled study of transcriptional regulation of de novo fatty acid (DNFA) production and de novo cholesterol synthesis (DNCS) in melanoma cell lines. This lipid-free ITS medium is able to support continuous proliferation of several melanoma cell lines that utilize DNL to support their lipid requirements. We show that the ITS medium stimulates gene transcription in support of both DNFA and DNCS, specifically mediated by SREBP1/2 in melanoma cells. We further found that the ITS medium promoted SREBP1 nuclear localization and occupancy on DNFA gene promoters. Our data show clear utility of this serum and lipid-free medium for melanoma cancer cell culture and lipid-related areas of investigation.


Asunto(s)
Ácidos Grasos/biosíntesis , Insulina/farmacología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Activación Transcripcional/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Colesterol/biosíntesis , Medios de Cultivo/química , Humanos , Melanoma/metabolismo , Melanoma/patología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Selenio/farmacología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Transferrina/farmacología
12.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955890

RESUMEN

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Asunto(s)
Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Receptor de Insulina/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor C1 de la Célula Huésped/antagonistas & inhibidores , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Insulina/metabolismo , Insulina/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Subunidades de Proteína/metabolismo , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor de Insulina/química , Transducción de Señal/efectos de los fármacos
13.
Nucleic Acid Ther ; 28(5): 273-284, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30133337

RESUMEN

The Ebola virus is a zoonotic pathogen that can cause severe hemorrhagic fever in humans, with up to 90% lethality. The deadly 2014 Ebola outbreak quickly made an unprecedented impact on human lives. While several vaccines and therapeutics are under development, current approaches contain several limitations, such as virus mutational escape, need for formulation or refrigeration, poor scalability, long lead-time, and high cost. To address these challenges, we developed locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) to target critical Ebola viral proteins and the human intracellular host protein Niemann-Pick C1 (NPC1), required for viral entry into infected cells. We generated noninfectious viral luciferase reporter assays to identify LNA ASOs that inhibit translation of Ebola viral proteins in vitro and in human cells. We demonstrated specific inhibition of key Ebola genes VP24 and nucleoprotein, which inhibit a proper immune response and promote Ebola virus replication, respectively. We also identified LNA ASOs targeting human host factor NPC1 and demonstrated reduced infection by chimeric vesicular stomatitis virus harboring the Ebola glycoprotein, which directly binds to NPC1 for viral infection. These results support further in vivo testing of LNA ASOs in infectious Ebola virus disease animal models as potential therapeutic modalities for treatment of Ebola.


Asunto(s)
Fiebre Hemorrágica Ebola/genética , Proteína Niemann-Pick C1/genética , Oligonucleótidos Antisentido/genética , Proteínas Virales/genética , Animales , Modelos Animales de Enfermedad , Ebolavirus/genética , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/terapia , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunidad Innata/genética , Ratones , Proteína Niemann-Pick C1/antagonistas & inhibidores , Nucleoproteínas/antagonistas & inhibidores , Nucleoproteínas/genética , Oligonucleótidos/genética , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/uso terapéutico , Primates/virología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/genética
14.
Trends Endocrinol Metab ; 29(10): 667-668, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29692333

RESUMEN

The miR-33 microRNAs (miRNAs) are crucial regulators of cholesterol/lipids, and may represent therapeutic targets for the treatment of atherosclerosis. A recent report by Price et al. showed that miR-33 knockout (KO) mice exhibit obesity, insulin resistance, and increased food intake, suggesting that metabolic regulation by miR-33 is more complex than was previously known.


Asunto(s)
Resistencia a la Insulina , MicroARNs , Adiposidad , Animales , Ratones , Ratones Noqueados , Obesidad , Expansión de Tejido
15.
J Biol Chem ; 291(37): 19651-60, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27471270

RESUMEN

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression by promoting degradation and/or repressing translation of specific target mRNAs. Several miRNAs have been identified that regulate the amplitude of the innate immune response by directly targeting Toll-like receptor (TLR) pathway members and/or cytokines. miR-33a and miR-33b (the latter present in primates but absent in rodents and lower species) are located in introns of the sterol regulatory element-binding protein (SREBP)-encoding genes and control cholesterol/lipid homeostasis in concert with their host gene products. These miRNAs regulate macrophage cholesterol by targeting the lipid efflux transporters ATP binding cassette (ABC)A1 and ABCG1. We and others have previously reported that Abca1(-/-) and Abcg1(-/-) macrophages have increased TLR proinflammatory responses due to augmented lipid raft cholesterol. Given this, we hypothesized that miR-33 would augment TLR signaling in macrophages via a raft cholesterol-dependent mechanism. Herein, we report that multiple TLR ligands down-regulate miR-33 in murine macrophages. In the case of lipopolysaccharide, this is a delayed, Toll/interleukin-1 receptor (TIR) domain-containing adapter-inducing interferon-ß-dependent response that also down-regulates Srebf-2, the host gene for miR-33. miR-33 augments macrophage lipid rafts and enhances proinflammatory cytokine induction and NF-κB activation by LPS. This occurs through an ABCA1- and ABCG1-dependent mechanism and is reversible by interventions upon raft cholesterol and by ABC transporter-inducing liver X receptor agonists. Taken together, these findings extend the purview of miR-33, identifying it as an indirect regulator of innate immunity that mediates bidirectional cross-talk between lipid homeostasis and inflammation.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/inmunología , Inmunidad Innata , Macrófagos/inmunología , Microdominios de Membrana/inmunología , MicroARNs/inmunología , Transportador 1 de Casete de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Animales , Microdominios de Membrana/genética , Ratones , Ratones Noqueados , MicroARNs/genética , Células RAW 264.7 , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/inmunología
16.
Biochim Biophys Acta ; 1861(12 Pt B): 2047-2052, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26968099

RESUMEN

In the past decade, microRNAs (miRNAs) have emerged as key regulators of circulating levels of lipoproteins. Specifically, recent work has uncovered the role of miRNAs in controlling the levels of atherogenic low-density lipoprotein LDL (LDL)-cholesterol by post-transcriptionally regulating genes involved in very low-density lipoprotein (VLDL) secretion, cholesterol biosynthesis, and hepatic LDL receptor (LDLR) expression. Interestingly, several of these miRNAs are located in genomic loci associated with abnormal levels of circulating lipids in humans. These findings reinforce the interest of targeting this subset of non-coding RNAs as potential therapeutic avenues for regulating plasma cholesterol and triglyceride (TAG) levels. In this review, we will discuss how these new miRNAs represent potential pre-disposition factors for cardiovascular disease (CVD), and putative therapeutic targets in patients with cardiometabolic disorders. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.


Asunto(s)
LDL-Colesterol/genética , MicroARNs/genética , Animales , Enfermedades Cardiovasculares/genética , Humanos , Enfermedades Metabólicas/genética , Receptores de LDL/genética , Triglicéridos/genética
17.
Nature ; 530(7591): 485-9, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26886795

RESUMEN

Eukaryotic transcription activators stimulate the expression of specific sets of target genes through recruitment of co-activators such as the RNA polymerase II-interacting Mediator complex. Aberrant function of transcription activators has been implicated in several diseases. However, therapeutic targeting efforts have been hampered by a lack of detailed molecular knowledge of the mechanisms of gene activation by disease-associated transcription activators. We previously identified an activator-targeted three-helix bundle KIX domain in the human MED15 Mediator subunit that is structurally conserved in Gal11/Med15 Mediator subunits in fungi. The Gal11/Med15 KIX domain engages pleiotropic drug resistance transcription factor (Pdr1) orthologues, which are key regulators of the multidrug resistance pathway in Saccharomyces cerevisiae and in the clinically important human pathogen Candida glabrata. The prevalence of C. glabrata is rising, partly owing to its low intrinsic susceptibility to azoles, the most widely used antifungal agent. Drug-resistant clinical isolates of C. glabrata most commonly contain point mutations in Pdr1 that render it constitutively active, suggesting that this transcriptional activation pathway represents a linchpin in C. glabrata multidrug resistance. Here we perform sequential biochemical and in vivo high-throughput screens to identify small-molecule inhibitors of the interaction of the C. glabrata Pdr1 activation domain with the C. glabrata Gal11A KIX domain. The lead compound (iKIX1) inhibits Pdr1-dependent gene activation and re-sensitizes drug-resistant C. glabrata to azole antifungals in vitro and in animal models for disseminated and urinary tract C. glabrata infection. Determining the NMR structure of the C. glabrata Gal11A KIX domain provides a detailed understanding of the molecular mechanism of Pdr1 gene activation and multidrug resistance inhibition by iKIX1. We have demonstrated the feasibility of small-molecule targeting of a transcription factor-binding site in Mediator as a novel therapeutic strategy in fungal infectious disease.


Asunto(s)
Antifúngicos/farmacología , Candida glabrata/efectos de los fármacos , Candida glabrata/metabolismo , Farmacorresistencia Fúngica/efectos de los fármacos , Proteínas Fúngicas/metabolismo , Complejo Mediador/metabolismo , Transactivadores/metabolismo , Animales , Sitios de Unión/efectos de los fármacos , Candida glabrata/genética , Candidiasis/tratamiento farmacológico , Candidiasis/microbiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Farmacorresistencia Fúngica Múltiple/efectos de los fármacos , Fluconazol/farmacología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Hidrazinas/farmacocinética , Hidrazinas/farmacología , Cetoconazol/farmacología , Complejo Mediador/química , Ratones , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Tiourea/análogos & derivados , Tiourea/farmacocinética , Tiourea/farmacología , Transactivadores/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
Nat Med ; 21(11): 1290-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501192

RESUMEN

Genome-wide association studies (GWASs) have linked genes to various pathological traits. However, the potential contribution of regulatory noncoding RNAs, such as microRNAs (miRNAs), to a genetic predisposition to pathological conditions has remained unclear. We leveraged GWAS meta-analysis data from >188,000 individuals to identify 69 miRNAs in physical proximity to single-nucleotide polymorphisms (SNPs) associated with abnormal levels of circulating lipids. Several of these miRNAs (miR-128-1, miR-148a, miR-130b, and miR-301b) control the expression of key proteins involved in cholesterol-lipoprotein trafficking, such as the low-density lipoprotein (LDL) receptor (LDLR) and the ATP-binding cassette A1 (ABCA1) cholesterol transporter. Consistent with human liver expression data and genetic links to abnormal blood lipid levels, overexpression and antisense targeting of miR-128-1 or miR-148a in high-fat diet-fed C57BL/6J and Apoe-null mice resulted in altered hepatic expression of proteins involved in lipid trafficking and metabolism, and in modulated levels of circulating lipoprotein-cholesterol and triglycerides. Taken together, these findings support the notion that altered expression of miRNAs may contribute to abnormal blood lipid levels, predisposing individuals to human cardiometabolic disorders.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Dieta Alta en Grasa , Dislipidemias/genética , MicroARNs/genética , Receptores de LDL/metabolismo , Triglicéridos/metabolismo , Animales , Apolipoproteínas E/genética , Colesterol/metabolismo , Estudio de Asociación del Genoma Completo , Homeostasis/genética , Humanos , Lipoproteínas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple
19.
Nat Med ; 21(11): 1280-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26437365

RESUMEN

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL cholesterol (LDL-C). Whereas the transcriptional regulation of LDLR is well characterized, the post-transcriptional mechanisms that govern LDLR expression are just beginning to emerge. Here we develop a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen we identified and characterized miR-148a as a negative regulator of LDLR expression and activity and defined a sterol regulatory element-binding protein 1 (SREBP1)-mediated pathway through which miR-148a regulates LDL-C uptake. In mice, inhibition of miR-148a increased hepatic LDLR expression and decreased plasma LDL-C. Moreover, we found that miR-148a regulates hepatic expression of ATP-binding cassette, subfamily A, member 1 (ABCA1) and circulating high-density lipoprotein cholesterol (HDL-C) levels in vivo. These studies uncover a role for miR-148a as a key regulator of hepatic LDL-C clearance through direct modulation of LDLR expression and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate an elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , MicroARNs/genética , Receptores de LDL/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Regulación de la Expresión Génica , Células Hep G2 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Receptores de LDL/metabolismo , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
20.
Nat Commun ; 6: 7505, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26106036

RESUMEN

Although BRCA1 function is essential for maintaining genomic integrity in all cell types, it is unclear why increased risk of cancer in individuals harbouring deleterious mutations in BRCA1 is restricted to only a select few tissues. Here we show that human mammary epithelial cells (HMECs) from BRCA1-mutation carriers (BRCA1(mut/+)) exhibit increased genomic instability and rapid telomere erosion in the absence of tumour-suppressor loss. Furthermore, we uncover a novel form of haploinsufficiency-induced senescence (HIS) specific to epithelial cells, which is triggered by pRb pathway activation rather than p53 induction. HIS and telomere erosion in HMECs correlate with misregulation of SIRT1 leading to increased levels of acetylated pRb as well as acetylated H4K16 both globally and at telomeric regions. These results identify a novel form of cellular senescence and provide a potential molecular basis for the rapid cell- and tissue- specific predisposition of breast cancer development associated with BRCA1 haploinsufficiency.


Asunto(s)
Senescencia Celular/genética , Células Epiteliales/metabolismo , Genes BRCA1 , Inestabilidad Genómica/genética , Haploinsuficiencia , Glándulas Mamarias Humanas/metabolismo , Acortamiento del Telómero/genética , Daño del ADN , Células Epiteliales/citología , Heterocigoto , Humanos , Glándulas Mamarias Humanas/citología , Mutación , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...