Try a new search

Format these results:

Searched for:

name:fernandez-hernando, carlos

school:SOM

Total Results:

112


Endothelial TGF-β signalling drives vascular inflammation and atherosclerosis

Chen, Pei-Yu; Qin, Lingfeng; Li, Guangxin; Wang, Zheng; Dahlman, James E; Malagon-Lopez, Jose; Gujja, Sharvari; Kauffman, Kevin J; Sun, Lele; Sun, Hongye; Zhang, Xinbo; Aryal, Binod; Canfran-Duque, Alberto; Liu, Rebecca; Kusters, Pascal; Sehgal, Alfica; Jiao, Yang; Anderson, Daniel G; Gulcher, Jeffrey; Fernandez-Hernando, Carlos; Lutgens, Esther; Schwartz, Martin A; Pober, Jordan S; Chittenden, Thomas W; Tellides, George; Simons, Michael
Atherosclerosis is a progressive vascular disease triggered by interplay between abnormal shear stress and endothelial lipid retention. A combination of these and, potentially, other factors leads to a chronic inflammatory response in the vessel wall, which is thought to be responsible for disease progression characterized by a buildup of atherosclerotic plaques. Yet molecular events responsible for maintenance of plaque inflammation and plaque growth have not been fully defined. Here we show that endothelial TGFβ signaling is one of the primary drivers of atherosclerosis-associated vascular inflammation. Inhibition of endothelial TGFβ signaling in hyperlipidemic mice reduces vessel wall inflammation and vascular permeability and leads to arrest of disease progression and regression of established lesions. These pro-inflammatory effects of endothelial TGFβ signaling are in stark contrast with its effects in other cell types and identify it as an important driver of atherosclerotic plaque growth and show the potential of cell-type specific therapeutic intervention aimed at control of this disease.
PMCID:6767930
PMID: 31572976
ISSN: 2522-5812
CID: 4118202

Specific Disruption of Abca1 Targeting Largely Mimics the Effects of miR-33 Knockout on Macrophage Cholesterol Efflux and Atherosclerotic Plaque Development

Price, Nathan L; Rotllan, Noemi; Zhang, Xinbo; Canfran-Duque, Alberto; Nottoli, Timothy; Suarez, Yajaira; Fernandez-Hernando, Carlos
RATIONALE/BACKGROUND:Inhibition of miR-33 reduces atherosclerotic plaque burden, but miR-33 deficient mice are predisposed to the development of obesity and metabolic dysfunction. The pro-atherogenic effects of miR-33 are thought to be in large part due to its repression of macrophage cholesterol efflux, through targeting of ATP Binding Cassette Subfamily A Member 1 ( Abca1). However, targeting of other factors may also be required for the beneficial effects of miR-33 and currently available approaches have not allowed researchers to determine the specific impact of individual miRNA target interactions in vivo. OBJECTIVE:In this work, we sought to determine how specific disruption of Abca1 targeting by miR-33 impacts macrophage cholesterol efflux and atherosclerotic plaque formation in vivo. METHODS AND RESULTS/RESULTS:mice had reduced atherosclerotic plaque formation, similar to mice transplanted with bone marrow from miR-33 knockout mice. CONCLUSIONS:Although the more pronounced phenotype of miR-33 deficient animals suggests that other targets may also play an important role, our data clearly demonstrate that repression of ABCA1 is primarily responsible for the pro-atherogenic effects of miR-33. This work shows for the first time that disruption of a single miRNA/target interaction can be sufficient to mimic the effects of miRNA deficiency on complex physiologic phenotypes in vivo and provides an approach by which to assess the impact of individual miRNA targets.
PMID: 30707082
ISSN: 1524-4571
CID: 3683772

Skeletal Muscle-Specific Deletion of MKP-1 Reveals a p38 MAPK/JNK/Akt Signaling Node That Regulates Obesity-Induced Insulin Resistance

Lawan, Ahmed; Min, Kisuk; Zhang, Lei; Canfran-Duque, Alberto; Jurczak, Michael J; Camporez, Joao Paulo G; Nie, Yaohui; Gavin, Timothy P; Shulman, Gerald I; Fernandez-Hernando, Carlos; Bennett, Anton M
Stress responses promote obesity and insulin resistance, in part, by activating the stress-responsive mitogen-activated protein kinases (MAPKs), p38 MAPK, and c-Jun NH2-terminal kinase (JNK). Stress also induces expression of MAPK phosphatase-1 (MKP-1), which inactivates both JNK and p38 MAPK. However, the equilibrium between JNK/p38 MAPK and MKP-1 signaling in the development of obesity and insulin resistance is unclear. Skeletal muscle is a major tissue involved in energy expenditure and glucose metabolism. In skeletal muscle, MKP-1 is upregulated in high-fat diet-fed mice and in skeletal muscle of obese humans. Mice lacking skeletal muscle expression of MKP-1 (MKP1-MKO) showed increased skeletal muscle p38 MAPK and JNK activities and were resistant to the development of diet-induced obesity. MKP1-MKO mice exhibited increased whole-body energy expenditure that was associated with elevated levels of myofiber-associated mitochondrial oxygen consumption. miR-21, a negative regulator of PTEN expression, was upregulated in skeletal muscle of MKP1-MKO mice, resulting in increased Akt activity consistent with enhanced insulin sensitivity. Our results demonstrate that skeletal muscle MKP-1 represents a critical signaling node through which inactivation of the p38 MAPK/JNK module promotes obesity and insulin resistance.
PMCID:5860856
PMID: 29317435
ISSN: 1939-327x
CID: 3064232

Macrophage deficiency of miR-21 promotes apoptosis, plaque necrosis, and vascular inflammation during atherogenesis

Canfran-Duque, Alberto; Rotllan, Noemi; Zhang, Xinbo; Fernandez-Fuertes, Marta; Ramirez-Hidalgo, Cristina; Araldi, Elisa; Daimiel, Lidia; Busto, Rebeca; Fernandez-Hernando, Carlos; Suarez, Yajaira
Atherosclerosis, the major cause of cardiovascular disease, is a chronic inflammatory disease characterized by the accumulation of lipids and inflammatory cells in the artery wall. Aberrant expression of microRNAs has been implicated in the pathophysiological processes underlying the progression of atherosclerosis. Here, we define the contribution of miR-21 in hematopoietic cells during atherogenesis. Interestingly, we found that miR-21 is the most abundant miRNA in macrophages and its absence results in accelerated atherosclerosis, plaque necrosis, and vascular inflammation. miR-21 expression influences foam cell formation, sensitivity to ER-stress-induced apoptosis, and phagocytic clearance capacity. Mechanistically, we discovered that the absence of miR-21 in macrophages increases the expression of the miR-21 target gene, MKK3, promoting the induction of p38-CHOP and JNK signaling. Both pathways enhance macrophage apoptosis and promote the post-translational degradation of ABCG1, a transporter that regulates cholesterol efflux in macrophages. Altogether, these findings reveal a major role for hematopoietic miR-21 in atherogenesis.
PMCID:5582411
PMID: 28674080
ISSN: 1757-4684
CID: 2617522

Engineered Microvasculature in PDMS Networks Using Endothelial Cells Derived from Human Induced Pluripotent Stem Cells

Sivarapatna, Amogh; Ghaedi, Mahboobe; Xiao, Yang; Han, Edward; Aryal, Binod; Zhou, Jing; Fernandez-Hernando, Carlos; Qyang, Yibing; Hirschi, Karen K; Niklason, Laura E
In this study, we used a polydimethylsiloxane (PDMS)-based platform for the generation of intact, perfusion-competent microvascular networks in vitro. COMSOL Multiphysics, a finite-element analysis and simulation software package, was used to obtain simulated velocity, pressure, and shear stress profiles. Transgene-free human induced pluripotent stem cells (hiPSCs) were differentiated into partially arterialized endothelial cells (hiPSC-ECs) in 5 d under completely chemically defined conditions, using the small molecule glycogen synthase kinase 3β inhibitor CHIR99021 and were thoroughly characterized for functionality and arterial-like marker expression. These cells, along with primary human umbilical vein endothelial cells (HUVECs), were seeded in the PDMS system to generate microvascular networks that were subjected to shear stress. Engineered microvessels had patent lumens and expressed VE-cadherin along their periphery. Shear stress caused by flowing medium increased the secretion of nitric oxide and caused endothelial cells s to align and to redistribute actin filaments parallel to the direction of the laminar flow. Shear stress also caused significant increases in gene expression for arterial markers Notch1 and EphrinB2 as well as antithrombotic markers Kruppel-like factor 2 (KLF-2)/4. These changes in response to shear stress in the microvascular platform were observed in hiPSC-EC microvessels but not in microvessels that were derived from HUVECs, which indicated that hiPSC-ECs may be more plastic in modulating their phenotype under flow than are HUVECs. Taken together, we demonstrate the feasibly of generating intact, engineered microvessels in vitro, which replicate some of the key biological features of native microvessels.
PMCID:5680973
PMID: 28901188
ISSN: 1555-3892
CID: 3071372

Lanosterol Modulates TLR4-Mediated Innate Immune Responses in Macrophages

Araldi, Elisa; Fernandez-Fuertes, Marta; Canfran-Duque, Alberto; Tang, Wenwen; Cline, Gary W; Madrigal-Matute, Julio; Pober, Jordan S; Lasuncion, Miguel A; Wu, Dianqing; Fernandez-Hernando, Carlos; Suarez, Yajaira
Macrophages perform critical functions in both innate immunity and cholesterol metabolism. Here, we report that activation of Toll-like receptor 4 (TLR4) in macrophages causes lanosterol, the first sterol intermediate in the cholesterol biosynthetic pathway, to accumulate. This effect is due to type I interferon (IFN)-dependent histone deacetylase 1 (HDAC1) transcriptional repression of lanosterol-14alpha-demethylase, the gene product of Cyp51A1. Lanosterol accumulation in macrophages, because of either treatment with ketoconazole or induced conditional disruption of Cyp51A1 in mouse macrophages in vitro, decreases IFNbeta-mediated signal transducer and activator of transcription (STAT)1-STAT2 activation and IFNbeta-stimulated gene expression. These effects translate into increased survival to endotoxemic shock by reducing cytokine secretion. In addition, lanosterol accumulation increases membrane fluidity and ROS production, thus potentiating phagocytosis and the ability to kill bacteria. This improves resistance of mice to Listeria monocytogenes infection by increasing bacterial clearance in the spleen and liver. Overall, our data indicate that lanosterol is an endogenous selective regulator of macrophage immunity.
PMCID:5553565
PMID: 28658622
ISSN: 2211-1247
CID: 2614152

miRNA regulation of white and brown adipose tissue differentiation and function

Price, Nathan L; Fernandez-Hernando, Carlos
Obesity and metabolic disorders are a major health concern in all developed countries and a primary focus of current medical research is to improve our understanding treatment of metabolic diseases. One avenue of research that has attracted a great deal of recent interest focuses upon understanding the role of miRNAs in the development of metabolic diseases. miRNAs have been shown to be dysregulated in a number of different tissues under conditions of obesity and insulin resistance, and have been demonstrated to be important regulators of a number of critical metabolic functions, including insulin secretion in the pancreas, lipid and glucose metabolism in the liver, and nutrient signaling in the hypothalamus. In this review we will focus on the important role of miRNAs in regulating the differentiation and function of white and brown adipose tissue and the potential importance of this for maintaining metabolic function and treating metabolic diseases. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernandez-Hernando and Yajaira Suarez.
PMCID:4987264
PMID: 26898181
ISSN: 0006-3002
CID: 2045622

miRNA regulation of LDL-cholesterol metabolism

Goedeke, Leigh; Wagschal, Alexandre; Fernandez-Hernando, Carlos; Naar, Anders M
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 Fernandez-Hernando and Yajaira Suarez.
PMCID:5375104
PMID: 26968099
ISSN: 0006-3002
CID: 2046922

Age-associated vascular inflammation promotes monocytosis during atherogenesis

Du, Wei; Wong, Christine; Song, Yang; Shen, Hua; Mori, Daniel; Rotllan, Noemi; Price, Nathan; Dobrian, Anca D; Meng, Hailong; Kleinstein, Steven H; Fernandez-Hernando, Carlos; Goldstein, Daniel R
Aging leads to a proinflammatory state within the vasculature without disease, yet whether this inflammatory state occurs during atherogenesis remains unclear. Here, we examined how aging impacts atherosclerosis using Ldlr(-/-) mice, an established murine model of atherosclerosis. We found that aged atherosclerotic Ldlr(-/-) mice exhibited enhanced atherogenesis within the aorta. Aging also led to increased LDL levels, elevated blood pressure on a low-fat diet, and insulin resistance after a high-fat diet (HFD). On a HFD, aging increased a monocytosis in the peripheral blood and enhanced macrophage accumulation within the aorta. When we conducted bone marrow transplant experiments, we found that stromal factors contributed to age-enhanced atherosclerosis. To delineate these stromal factors, we determined that the vasculature exhibited an age-enhanced inflammatory response consisting of elevated production of CCL-2, osteopontin, and IL-6 during atherogenesis. In addition, in vitro cultures showed that aging enhanced the production of osteopontin by vascular smooth muscle cells. Functionally, aged atherosclerotic aortas displayed higher monocyte chemotaxis than young aortas. Hence, our study has revealed that aging induces metabolic dysfunction and enhances vascular inflammation to promote a peripheral monocytosis and macrophage accumulation within the atherosclerotic aorta.
PMCID:4933655
PMID: 27135421
ISSN: 1474-9726
CID: 3102792

ANGPTL4 deficiency in haematopoietic cells promotes monocyte expansion and atherosclerosis progression

Aryal, Binod; Rotllan, Noemi; Araldi, Elisa; Ramirez, Cristina M; He, Shun; Chousterman, Benjamin G; Fenn, Ashley M; Wanschel, Amarylis; Madrigal-Matute, Julio; Warrier, Nikhil; Martin-Ventura, Jose L; Swirski, Filip K; Suarez, Yajaira; Fernandez-Hernando, Carlos
Lipid accumulation in macrophages has profound effects on macrophage gene expression and contributes to the development of atherosclerosis. Here, we report that angiopoietin-like protein 4 (ANGPTL4) is the most highly upregulated gene in foamy macrophages and it's absence in haematopoietic cells results in larger atherosclerotic plaques, characterized by bigger necrotic core areas and increased macrophage apoptosis. Furthermore, hyperlipidemic mice deficient in haematopoietic ANGPTL4 have higher blood leukocyte counts, which is associated with an increase in the common myeloid progenitor (CMP) population. ANGPTL4-deficient CMPs have higher lipid raft content, are more proliferative and less apoptotic compared with the wild-type (WT) CMPs. Finally, we observe that ANGPTL4 deficiency in macrophages promotes foam cell formation by enhancing CD36 expression and reducing ABCA1 localization in the cell surface. Altogether, these findings demonstrate that haematopoietic ANGPTL4 deficiency increases atherogenesis through regulating myeloid progenitor cell expansion and differentiation, foam cell formation and vascular inflammation.
PMCID:4974469
PMID: 27460411
ISSN: 2041-1723
CID: 2191152