Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Noemi Rotllan is active.

Publication


Featured researches published by Noemi Rotllan.


PLOS ONE | 2011

Statins Promote the Regression of Atherosclerosis via Activation of the CCR7-Dependent Emigration Pathway in Macrophages

Jonathan E. Feig; Yueting Shang; Noemi Rotllan; Yuliya Vengrenyuk; Chaowei Wu; Raanan Shamir; Inés Pineda Torra; Carlos Fernández-Hernando; Edward A. Fisher; Michael J. Garabedian

HMG-CoA reductase inhibitors (statins) decrease atherosclerosis by lowering low-density-lipoprotein cholesterol. Statins are also thought to have additional anti-atherogenic properties, yet defining these non-conventional modes of statin action remains incomplete. We have previously developed a novel mouse transplant model of atherosclerosis regression in which aortic segments from diseased donors are placed into normolipidemic recipients. With this model, we demonstrated the rapid loss of CD68+ cells (mainly macrophages) in plaques through the induction of a chemokine receptor CCR7-dependent emigration process. Because the human and mouse CCR7 promoter contain Sterol Response Elements (SREs), we hypothesized that Sterol Regulatory Element Binding Proteins (SREBPs) are involved in increasing CCR7 expression and through this mechanism, statins would promote CD68+ cell emigration from plaques. We examined whether statin activation of the SREBP pathway in vivo would induce CCR7 expression and promote macrophage emigration from plaques. We found that western diet-fed apoE(-/-) mice treated with either atorvastatin or rosuvastatin led to a substantial reduction in the CD68+ cell content in the plaques despite continued hyperlipidemia. We also observed a significant increase in CCR7 mRNA in CD68+ cells from both the atorvastatin and rosuvastatin treated mice associated with emigration of CD68+ cells from plaques. Importantly, CCR7(-/-)/apoE(-/-) double knockout mice failed to display a reduction in CD68+ cell content upon statin treatment. Statins also affected the recruitment of transcriptional regulatory proteins and the organization of the chromatin at the CCR7 promoter to increase the transcriptional activity. Statins promote the beneficial remodeling of plaques in diseased mouse arteries through the stimulation of the CCR7 emigration pathway in macrophages. Therefore, statins may exhibit some of their clinical benefits by not only retarding the progression of atherosclerosis, but also accelerating its regression.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Therapeutic Silencing of MicroRNA-33 Inhibits the Progression of Atherosclerosis in Ldlr−/− Mice—Brief Report

Noemi Rotllan; Cristina M. Ramírez; Binod Aryal; Christine Esau; Carlos Fernández-Hernando

Objective—To study the efficacy of anti–miRNA-33 therapy on the progression of atherosclerosis. Approach and Results—Ldlr−/− mice were injected subcutaneously with PBS, control, or anti–miR-33 oligonucleotides weekly and fed a Western diet for 12 weeks. At the end of treatment, the expression of miR-33 target genes was increased in the liver and aorta, demonstrating effective inhibition of miR-33 function. Interestingly, plasma high-density lipoprotein (HDL)-cholesterol was significantly increased in anti–miR-33-treated mice but only when they were fed a chow diet. However, HDL isolated from anti–miR-33-treated mice showed an increase cholesterol efflux capacity compared with HDL isolated from nontargeting oligonucleotide-treated mice. Analysis of atherosclerosis revealed a significant reduction of plaque size and macrophage content in mice receiving anti–miR-33. In contrast, no differences in collagen content and necrotic areas were observed among the 3 groups. Conclusions—Long-term anti–miR-33 therapy significantly reduces the progression of atherosclerosis and improves HDL functionality. The antiatherogenic effect is independent of plasma HDL-cholesterol levels.


The FASEB Journal | 2013

Cardiovascular dysregulation of miR-17-92 causes a lethal hypertrophic cardiomyopathy and arrhythmogenesis

Laura S. Danielson; David S. Park; Noemi Rotllan; Aránzazu Chamorro-Jorganes; Maria V. Guijarro; Carlos Fernández-Hernando; Glenn I. Fishman; Colin K.L. Phoon; Eva Hernando

MicroRNA cluster miR‐17‐92 has been implicated in cardiovascular development and function, yet its precise mechanisms of action in these contexts are uncertain. This study aimed to investigate the role of miR‐17‐92 in morphogenesis and function of cardiac and smooth muscle tissues. To do so, a mouse model of conditional overexpression of miR‐17‐92 in cardiac and smooth muscle tissues was generated. Extensive cardiac functional studies identified a dose‐dependent induction of dilated, hypertrophic cardiomyopathy, and arrhythmia inducibility in transgenic animals, which correlated with premature mortality (98.3±42.5 d, P<0.0001). Expression analyses revealed the abundance of Pten transcript, a known miR‐17‐92 target, to be inversely correlated with miR‐17‐92 expression levels and heart size. In addition, we demonstrated through 3′‐UTR luciferase assays and expression analyses that Connexin43 (Cx43) is a novel direct target of miR‐19a/b and its expression is suppressed in transgenic hearts. Taken together, these data demonstrate that dysregulated expression of miR‐17‐92 during cardiovascular morphogenesis results in a lethal cardiomyopathy, possibly in part through direct repression of Pten and Cx43. This study highlights the importance of miR‐17‐92 in both normal and pathological functions of the heart, and provides a model that may serve as a useful platform to test novel antiarrhythmic therapeutics.—Danielson, L. S., Park, D. S., Rotllan, N., Chamorro‐Jorganes, A., Guijarro, M. V., Fernandez‐Hernando, C., Fishman, G. I., Phoon, C. K. L., Hernando, E. Cardiovascular dysregulation of miR‐17‐92 causes a lethal hypertrophic cardiomyopathy and arrhythmogenesis. FASEB J. 27, 1460–1467 (2013). www.fasebj.org


Circulation Research | 2013

Control of Cholesterol Metabolism and Plasma High-Density Lipoprotein Levels by microRNA-144Novelty and Significance

Cristina M. Ramírez; Noemi Rotllan; Alexander V. Vlassov; Alberto Dávalos; Mu Li; Leigh Goedeke; Juan F. Aranda; Daniel Cirera-Salinas; Elisa Araldi; Alessandro G. Salerno; Amarylis Wanschel; Jiri Zavadil; Antonio Castrillo; Jungsu Kim; Yajaira Suárez; Carlos Fernández-Hernando

Rationale: Foam cell formation because of excessive accumulation of cholesterol by macrophages is a pathological hallmark of atherosclerosis, the major cause of morbidity and mortality in Western societies. Liver X nuclear receptors (LXRs) regulate the expression of the adenosine triphosphate–binding cassette (ABC) transporters, including adenosine triphosphate–binding cassette transporter A1 (ABCA1) and adenosine triphosphate–binding cassette transporter G1 (ABCG1). ABCA1 and ABCG1 facilitate the efflux of cholesterol from macrophages and regulate high-density lipoprotein (HDL) biogenesis. Increasing evidence supports the role of microRNA (miRNAs) in regulating cholesterol metabolism through ABC transporters. Objective: We aimed to identify novel miRNAs that regulate cholesterol metabolism in macrophages stimulated with LXR agonists. Methods and Results: To map the miRNA expression signature of macrophages stimulated with LXR agonists, we performed an miRNA profiling microarray analysis in primary mouse peritoneal macrophages stimulated with LXR ligands. We report that LXR ligands increase miR-144 expression in macrophages and mouse livers. Overexpression of miR-144 reduces ABCA1 expression and attenuates cholesterol efflux to apolipoproteinA1 in macrophages. Delivery of miR-144 oligonucleotides to mice attenuates ABCA1 expression in the liver, reducing HDL levels. Conversely, silencing of miR-144 in mice increases the expression of ABCA1 and plasma HDL levels. Thus, miR-144 seems to regulate both macrophage cholesterol efflux and HDL biogenesis in the liver. Conclusions: miR-144 regulates cholesterol metabolism via suppressing ABCA1 expression and modulation of miRNAs may represent a potential therapeutical intervention for treating dyslipidemia and atherosclerotic vascular disease.


Molecular and Cellular Biology | 2013

MicroRNA 33 Regulates Glucose Metabolism

Cristina M. Ramírez; Leigh Goedeke; Noemi Rotllan; Je-Hyun Yoon; Daniel Cirera-Salinas; Julie A. Mattison; Yajaira Suárez; Rafael de Cabo; Myriam Gorospe; Carlos Fernández-Hernando

ABSTRACT Metabolic diseases are characterized by the failure of regulatory genes or proteins to effectively orchestrate specific pathways involved in the control of many biological processes. In addition to the classical regulators, recent discoveries have shown the remarkable role of small noncoding RNAs (microRNAs [miRNAs]) in the posttranscriptional regulation of gene expression. In this regard, we have recently demonstrated that miR-33a and miR33b, intronic miRNAs located within the sterol regulatory element-binding protein (SREBP) genes, regulate lipid metabolism in concert with their host genes. Here, we show that miR-33b also cooperates with SREBP1 in regulating glucose metabolism by targeting phosphoenolpyruvate carboxykinase (PCK1) and glucose-6-phosphatase (G6PC), key regulatory enzymes of hepatic gluconeogenesis. Overexpression of miR-33b in human hepatic cells inhibits PCK1 and G6PC expression, leading to a significant reduction of glucose production. Importantly, hepatic SREBP1c/miR-33b levels correlate inversely with the expression of PCK1 and G6PC upon glucose infusion in rhesus monkeys. Taken together, these results suggest that miR-33b works in concert with its host gene to ensure a fine-tuned regulation of lipid and glucose homeostasis, highlighting the clinical potential of miR-33a/b as novel therapeutic targets for a range of metabolic diseases.


Current Atherosclerosis Reports | 2013

MicroRNAs and Atherosclerosis

Noemi Rotllan; Juan F. Aranda; Carlos Fernández-Hernando

MicroRNAs (miRNAs) are small, ~22 nucleotide (nt) sequences of RNA that regulate gene expression at posttranscriptional level. These endogenous gene expression inhibitors were primarily described in cancer but recent exciting findings have also demonstrated a key role in cardiovascular diseases (CVDs), including atherosclerosis. MiRNAs control endothelial cell (EC), vascular smooth muscle cell (VSMC), and macrophage functions, and thereby regulate the progression of atherosclerosis. MiRNA expression is modulated by different stimuli involved in every stage of atherosclerosis, and conversely miRNAs modulates several pathways implicated in plaque development such as cholesterol metabolism. In the present review, we focus on the importance of miRNAs in atherosclerosis, and we further discuss their potential use as biomarkers and therapeutic targets in CVDs.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2005

Overexpression of Human Apolipoprotein A-II in Transgenic Mice Does Not Impair Macrophage-Specific Reverse Cholesterol Transport In Vivo

Noemi Rotllan; Vicent Ribas; Laura Calpe-Berdiel; Jesús M. Martín-Campos; Francisco Blanco-Vaca; Joan Carles Escolà-Gil

BACKGROUND Overexpression of human apolipoprotein (apo) A-II in transgenic mice induces high-density lipoprotein (HDL) deficiency, and increased atherosclerosis susceptibility only when fed an atherogenic diet. This may, in part, be caused by impairment in reverse cholesterol transport (RCT). METHODS AND RESULTS [3H]cholesterol-labeled macrophages were injected intraperitoneally into mice maintained on a chow diet or an atherogenic diet. Plasma [3H]cholesterol did not differ from human apoA-II transgenic and control mice at 24 or 48 hours after the label injection. On the chow diet, human apoA-II transgenic mice presented increased [3H]cholesterol in liver (1.3-fold) and feces (6-fold) compared with control mice (P<0.05). The magnitude of macrophage-specific RCT did not differ between transgenic and control mice fed the atherogenic diet. CONCLUSIONS Human apoA-II maintains effective RCT from macrophages to feces in vivo despite an HDL deficiency. These findings suggest that the increased atherosclerotic lesions observed in apoA-II transgenic mice fed an atherogenic diet are not caused by impairment in macrophage-specific RCT.


Nature Medicine | 2015

MicroRNA-148a regulates LDL receptor and ABCA1 expression to control circulating lipoprotein levels.

Leigh Goedeke; Noemi Rotllan; Alberto Canfrán-Duque; Juan F. Aranda; Cristina M. Ramírez; Elisa Araldi; Chin Sheng Lin; Norma N. Anderson; Alexandre Wagschal; Rafael de Cabo; Jay D. Horton; Miguel A. Lasunción; Anders M. Näär; Yajaira Suárez; Carlos Fernández-Hernando

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL-cholesterol (LDL-C). While the transcriptional regulation of LDLR is well-characterized, the post-transcriptional mechanisms which govern LDLR expression are just beginning to emerge. Here, we developed a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen, we characterize miR-148a as a negative regulator of LDLR expression and activity, and define a novel SREBP1-mediated pathway by which miR-148a regulates LDL-C uptake. Importantly, inhibition of miR-148a increases hepatic LDLR expression and decreases plasma LDL-C in vivo. We also provide evidence that miR-148a regulates hepatic ABCA1 expression and circulating HDL-C levels. Collectively, these studies uncover miR-148a as an important regulator of hepatic LDL-C clearance through direct regulation of LDLR expression, and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate the elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Human Apolipoprotein A-II Determines Plasma Triglycerides by Regulating Lipoprotein Lipase Activity and High-Density Lipoprotein Proteome

Josep Julve; Joan Carles Escolà-Gil; Noemi Rotllan; Catherine Fievet; Emmanuelle Vallez; Carolina de la Torre; Vicent Ribas; John H. Sloan; Francisco Blanco-Vaca

Introduction—Apolipoprotein (apo) A-II is the second most abundant high-density lipoprotein (HDL) apolipoprotein. We assessed the mechanism involved in the altered postprandial triglyceride-rich lipoprotein metabolism of female human apoA-II-transgenic mice (hapoA-II-Tg mice), which results in up to an 11-fold increase in plasma triglyceride concentration. The relationships between apoA-II, HDL composition, and lipoprotein lipase (LPL) activity were also analyzed in a group of normolipidemic women. Methods and Results—Triglyceride-rich lipoprotein catabolism was decreased in hapoA-II-Tg mice compared to control mice. This suggests that hapoA-II, which was mainly associated with HDL during fasting and postprandially, impairs triglyceride-rich lipoprotein lipolysis. HDL isolated from hapoA-II-Tg mice impaired bovine LPL activity. Two-dimensional gel electrophoresis, mass spectrometry, and immunonephelometry identified a marked deficiency in the HDL content of apoA-I, apoC-III, and apoE in these mice. In normolipidemic women, apoA-II concentration was directly correlated with plasma triglyceride and inversely correlated with the HDL-apoC-II+apoE/apoC-III ratio. HDL-mediated induction of LPL activity was inversely correlated with apoA-II and directly correlated with the HDL-apoC-II+apoE/apoC-III ratio. Purified hapoA-II displaced apoC-II, apoC-III, and apoE from human HDL2. Human HDL3 was, compared to HDL2, enriched in apoA-II but poorer in apoC-II, apoC-III, and apoE. Conclusion—ApoA-II plays a crucial role in triglyceride catabolism by regulating LPL activity, at least in part, through HDL proteome modulation.


Atherosclerosis | 2009

In vivo macrophage-specific RCT and antioxidant and antiinflammatory HDL activity measurements: New tools for predicting HDL atheroprotection

Joan Carles Escolà-Gil; Noemi Rotllan; Josep Julve; Francisco Blanco-Vaca

The beneficial therapeutic effects of raising HDL cholesterol are proving difficult to confirm in humans. The evaluation of antiatherogenic functions of HDL is an important area of research which includes the role of HDL in reverse cholesterol transport (RCT), especially macrophage-specific RCT, and its antioxidant and antiinflammatory roles. The antioxidant and antiinflammatory functions of HDL can be assessed using cell-free and cell-based assays. Also, a new approach was developed to measure RCT from labeled-cholesterol macrophages to liver and feces of mice. Studies in genetically engineered animals indicate that these major HDL antiatherogenic functions are better predictors of atherosclerosis susceptibility than HDL cholesterol or total RCT. Thus, functional testing of the antiatherogenic functions of HDL in experimental animal models may facilitate the development of new strategies for the prevention and treatment of atherosclerosis.

Collaboration


Dive into the Noemi Rotllan's collaboration.

Top Co-Authors

Avatar

Francisco Blanco-Vaca

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joan Carles Escolà-Gil

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cristina M. Ramírez

University of Las Palmas de Gran Canaria

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juan F. Aranda

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge