Network


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

Hotspot


Dive into the research topics where Rajendra K. Tangirala is active.

Publication


Featured researches published by Rajendra K. Tangirala.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Identification of macrophage liver X receptors as inhibitors of atherosclerosis

Rajendra K. Tangirala; Eric D. Bischoff; Sean B. Joseph; Brandee L. Wagner; Robert Walczak; Bryan A. Laffitte; Chris L. Daige; Diane Thomas; Richard A. Heyman; David J. Mangelsdorf; Xuping Wang; Aldons J. Lusis; Peter Tontonoz; Ira G. Schulman

Recent studies have identified the liver X receptors (LXRα and LXRβ) as important regulators of cholesterol metabolism and transport. LXRs control transcription of genes critical to a range of biological functions including regulation of high density lipoprotein cholesterol metabolism, hepatic cholesterol catabolism, and intestinal sterol absorption. Although LXR activity has been proposed to be critical for physiologic lipid metabolism and transport, direct evidence linking LXR signaling pathways to the pathogenesis of cardiovascular disease has yet to be established. In this study bone marrow transplantations were used to selectively eliminate macrophage LXR expression in the context of murine models of atherosclerosis. Our results demonstrate that LXRs are endogenous inhibitors of atherogenesis. Additionally, elimination of LXR activity in bone marrow-derived cells mimics many aspects of Tangier disease, a human high density lipoprotein deficiency, including aberrant regulation of cholesterol transporter expression, lipid accumulation in macrophages, splenomegaly, and increased atherosclerosis. These results identify LXRs as targets for intervention in cardiovascular disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2004

Macrophage Liver X Receptor Is Required for Antiatherogenic Activity of LXR Agonists

Nancy Levin; Eric D. Bischoff; Chris L. Daige; Diane Thomas; Calvin T. Vu; Richard A. Heyman; Rajendra K. Tangirala; Ira G. Schulman

Objective— Complications of atherosclerotic cardiovascular disease due to elevated blood cholesterol levels are the major cause of death in the Western world. The liver X receptors, LXR&agr; and LXR&bgr; (LXRs), are ligand-dependent transcription factors that act as cholesterol sensors and coordinately control transcription of genes involved in cholesterol and lipid homeostasis as well as macrophage inflammatory gene expression. LXRs regulate cholesterol balance through activation of ATP-binding cassette transporters that promote cholesterol transport and excretion from the liver, intestine, and macrophage. Although LXR agonists are known to delay progression of atherosclerosis in mouse models, their ability to abrogate preexisting cardiovascular disease by inducing regression and stabilization of established atherosclerotic lesions has not been addressed. Methods and Results— We demonstrate that LXR agonist treatment increases ATP-binding cassette transporter expression within preexisting atherosclerotic lesions, resulting in regression of these lesions as well as remodeling from vulnerable to stable lesions and a reduction in macrophage content. Further, using macrophage-selective LXR-deficient mice created by bone marrow transplantation, we provide the first evidence that macrophage LXR expression is necessary for the atheroprotective actions of an LXR agonist. Conclusions— These data substantiate that drugs targeting macrophage LXR activity may offer therapeutic benefit in the treatment of atherosclerotic cardiovascular disease.


Journal of Clinical Investigation | 2007

Ligand activation of LXRβ reverses atherosclerosis and cellular cholesterol overload in mice lacking LXRα and apoE

Michelle N. Bradley; Cynthia Hong; Mingyi Chen; Sean B. Joseph; Damien C. Wilpitz; Xuping Wang; Aldons J. Lusis; Allan J. Collins; Willa A. Hseuh; Jon L. Collins; Rajendra K. Tangirala; Peter Tontonoz

Liver X receptors (LXRs) α and β are transcriptional regulators of cholesterol homeostasis and potential targets for the development of antiatherosclerosis drugs. However, the specific roles of individual LXR isotypes in atherosclerosis and the pharmacological effects of synthetic agonists remain unclear. Previous work has shown that mice lacking LXRα accumulate cholesterol in the liver but not in peripheral tissues. In striking contrast, we demonstrate here that LXRα–/–apoE–/– mice exhibit extreme cholesterol accumulation in peripheral tissues, a dramatic increase in whole-body cholesterol burden, and accelerated atherosclerosis. The phenotype of these mice suggests that the level of LXR pathway activation in macrophages achieved by LXRβ and endogenous ligand is unable to maintain homeostasis in the setting of hypercholesterolemia. Surprisingly, however, a highly efficacious synthetic agonist was able to compensate for the loss of LXRα. Treatment of LXRα–/–apoE–/– mice with synthetic LXR ligand ameliorates the cholesterol overload phenotype and reduces atherosclerosis. These observations indicate that LXRα has an essential role in maintaining peripheral cholesterol homeostasis in the context of hypercholesterolemia and provide in vivo support for drug development strategies targeting LXRβ.


Proceedings of the National Academy of Sciences of the United States of America | 2008

PPARδ-mediated antiinflammatory mechanisms inhibit angiotensin II-accelerated atherosclerosis

Yasunori Takata; Joey Liu; Fen Yin; Alan R. Collins; Christopher J. Lyon; Chih-Hao Lee; Annette R. Atkins; Michael Downes; Grant D. Barish; Ronald M. Evans; Willa A. Hsueh; Rajendra K. Tangirala

Activation of the nuclear hormone receptor peroxisome proliferator-activated receptor δ (PPARδ) has been shown to improve insulin resistance, adiposity, and plasma HDL levels. However, its antiatherogenic role remains controversial. Here we report atheroprotective effects of PPARδ activation in a model of angiotensin II (AngII)-accelerated atherosclerosis, characterized by increased vascular inflammation related to repression of an antiinflammatory corepressor, B cell lymphoma-6 (Bcl-6), and the regulators of G protein-coupled signaling (RGS) proteins RGS4 and RGS5. In this model, administration of the PPARδ agonist GW0742 (1 or 10 mg/kg) substantially attenuated AngII-accelerated atherosclerosis without altering blood pressure and increased vascular expression of Bcl-6, RGS4, and RGS5, which was associated with suppression of inflammatory and atherogenic gene expression in the artery. In vitro studies demonstrated similar changes in AngII-treated macrophages: PPARδ activation increased both total and free Bcl-6 levels and inhibited AngII activation of MAP kinases, p38, and ERK1/2. These studies uncover crucial proinflammatory mechanisms of AngII and highlight actions of PPARδ activation to inhibit AngII signaling, which is atheroprotective.


Genes & Development | 2010

Bcl-6 and NF-κB cistromes mediate opposing regulation of the innate immune response

Grant D. Barish; Ruth T. Yu; Malith S. Karunasiri; Corinne B. Ocampo; Jesse Dixon; Christopher Benner; Alexander L. Dent; Rajendra K. Tangirala; Ronald M. Evans

In the macrophage, toll-like receptors (TLRs) are key sensors that trigger signaling cascades to activate inflammatory programs via the NF-κB gene network. However, the genomic network targeted by TLR/NF-κB activation and the molecular basis by which it is restrained to terminate activation and re-establish quiescence is poorly understood. Here, using chromatin immunoprecipitation sequencing (ChIP-seq), we define the NF-κB cistrome, which is comprised of 31,070 cis-acting binding sites responsive to lipopolysaccharide (LPS)-induced signaling. In addition, we demonstrate that the transcriptional repressor B-cell lymphoma 6 (Bcl-6) regulates nearly a third of the Tlr4-regulated transcriptome, and that 90% of the Bcl-6 cistrome is collapsed following Tlr4 activation. Bcl-6-deficient macrophages are acutely hypersensitive to LPS and, using comparative ChIP-seq analyses, we found that the Bcl-6 and NF-κB cistromes intersect, within nucleosomal distance, at nearly half of Bcl-6-binding sites in stimulated macrophages to promote opposing epigenetic modifications of the local chromatin. These results reveal a genomic strategy for controlling the innate immune response in which repressive and inductive cistromes establish a dynamic balance between macrophage quiescence and activation via epigenetically marked cis-regulatory elements.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1998

Chemokine Receptor CCR2 Expression and Monocyte Chemoattractant Protein-1–Mediated Chemotaxis in Human Monocytes A Regulatory Role for Plasma LDL

Ki Hoon Han; Rajendra K. Tangirala; Simone R. Green; Oswald Quehenberger

The subendothelial accumulation of macrophage-derived foam cells is one of the hallmarks of atherosclerosis. The recruitment of monocytes to the intima requires the interaction of locally produced chemokines with specific cell surface receptors, including the receptor (CCR2) for monocyte chemoattractant protein-1 (MCP-1). We have previously reported that monocyte CCR2 gene expression and function are effectively downregulated by proinflammatory cytokines. In this study we identified low density lipoprotein (LDL) as a positive regulator of CCR2 expression. Monocyte CCR2 expression was dramatically increased in hypercholesterolemic patients compared with normocholesterolemic controls. Similarly, incubation of human THP-1 monocytes with LDL induced a rapid increase in CCR2 mRNA and protein. By 24 hours the number of cell surface receptors was doubled, causing a 3-fold increase in the chemotactic response to MCP-1. The increase in CCR2 expression and chemotaxis was promoted by native LDL but not by oxidized LDL. Oxidized LDL rapidly downregulated CCR2 expression, whereas reductively methylated LDL, which does not bind to the LDL receptor, had only modest effects on CCR2 expression. A neutralizing anti-LDL receptor antibody prevented the effect of LDL, suggesting that binding and internalization of LDL were essential for CCR2 upregulation. The induction of CCR2 expression appeared to be mediated by LDL-derived cholesterol, because cells treated with free cholesterol also showed increased CCR2 expression. These data suggest that elevated plasma LDL levels in conditions such as hypercholesterolemia enhance monocyte CCR2 expression and chemotactic response and potentially contribute to increased monocyte recruitment to the vessel wall in chronic inflammation and atherogenesis.


Circulation Research | 2009

Age-Accelerated Atherosclerosis Correlates With Failure to Upregulate Antioxidant Genes

Alan R. Collins; Christopher J. Lyon; Xuefeng Xia; Joey Z. Liu; Rajendra K. Tangirala; Fen Yin; Rima Boyadjian; Alfiya Bikineyeva; Domenico Praticò; David G. Harrison; Willa A. Hsueh

Excess food intake leads to obesity and diabetes, both of which are well-known independent risk factors for atherosclerosis, and both of which are growing epidemics in an aging population. We hypothesized that aging enhances the metabolic and vascular effects of high fat diet (HFD) and therefore examined the effect of age on atherosclerosis and insulin resistance in lipoprotein receptor knockout (LDLR−/−) mice. We found that 12-month-old (middle-aged) LDLR−/− mice developed substantially worse metabolic syndrome, diabetes, and atherosclerosis than 3-month-old (young) LDLR−/− mice when both were fed HFD for 3 months, despite similar elevations in total cholesterol levels. Microarray analyses were performed to analyze the mechanism responsible for the marked acceleration of atherosclerosis in middle-aged mice. Chow-fed middle-aged mice had greater aortic expression of multiple antioxidant genes than chow-fed young mice, including glutathione peroxidase-1 and -4, catalase, superoxide dismutase-2, and uncoupling protein-2. Aortic expression of these enzymes markedly increased in young mice fed HFD but decreased or only modestly increased in middle-aged mice fed HFD, despite the fact that systemic oxidative stress and vascular reactive oxygen species generation, measured by plasma F2α isoprostane concentration (systemic) and dihydroethidium conversion and p47phox expression (vascular), were greater in middle-aged mice fed HFD. Thus, the mechanism for the accelerated vascular injury in older LDLR−/− mice was likely the profound inability to mount an antioxidant response. This effect was related to a decrease in vascular expression of 2 key transcriptional pathways regulating the antioxidant response, DJ-1 and forkhead box, subgroup O family (FOXOs). Treatment of middle-aged mice fed HFD with the antioxidant apocynin attenuated atherosclerosis, whereas treatment with the insulin sensitizer rosiglitazone attenuated both metabolic syndrome and atherosclerosis. Both treatments decreased oxidative stress. A novel effect of rosiglitazone was to increase expression of Nrf2 (nuclear factor [erythroid-derived 2]-like 2), a downstream target of DJ-1 contributing to enhanced expression of vascular antioxidant enzymes. This investigation underscores the role of oxidative stress when multiple atherosclerotic risk factors, particularly aging, converge on the vessel wall and emphasizes the need to develop effective strategies to inhibit oxidative stress to protect aging vasculature.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2006

Impaired Development of Atherosclerosis in Hyperlipidemic Ldlr−/− and ApoE−/− Mice Transplanted With Abcg1−/− Bone Marrow

Ángel Baldán; Liming Pei; Richard T. Lee; Paul T. Tarr; Rajendra K. Tangirala; Michael M. Weinstein; Joy S. Frank; Andrew C. Li; Peter Tontonoz; Peter A. Edwards

Objective—The lungs of Abcg1−/− mice accumulate macrophage foam cells that contain high levels of unesterified and esterified cholesterol, consistent with a role for ABCG1 in facilitating the efflux of cholesterol from macrophages to high-density lipoprotein (HDL) and other exogenous sterol acceptors. Based on these observations, we investigated whether loss of ABCG1 affects foam cell deposition in the artery wall and the development of atherosclerosis. Methods and Results—Bone marrow from wild-type or Abcg1−/− mice was transplanted into Ldlr−/− or ApoE−/− mice. After administration of a high-fat/high-cholesterol diet, plasma and tissue lipid levels and atherosclerotic lesion size were quantified and compared. Surprisingly, transplantation of Abcg1−/− bone marrow cells resulted in a significant reduction in lesion size in both mouse models, despite the fact that lipid levels increased in the lung, spleen, and kidney. Lesions of Ldlr−/− mice transplanted with Abcg1−/− cells contained increased numbers of apoptotic cells. Consistent with this observation, in vitro studies demonstrated that Abcg1−/− macrophages were more susceptible to oxidized low-density lipoprotein (ox-LDL)-dependent apoptosis than Abcg1+/+ cells. Conclusions—Diet-induced atherosclerosis is impaired when atherosclerotic-susceptible mice are transplanted with Abcg1−/− bone marrow. The demonstration that Abcg1−/− macrophages undergo accelerated apoptosis provides a mechanism to explain the decrease in the atherosclerotic lesions.


Journal of Biological Chemistry | 1997

Regulation of expression of the human monocyte chemotactic protein-1 receptor (hCCR2) by cytokines.

Rajendra K. Tangirala; Koji Murao; Oswald Quehenberger

Monocytes enter the subendothelial space in response to a variety of chemotactic agents, notably including monocyte chemotactic protein-1 (MCP-1). To better understand the role of the human MCP-1 receptor (hCCR2) in monocyte recruitment, we have examined the effects of cytokines on expression of the receptor gene by ligand binding and Northern blot analysis. THP-1 cells expressed on average about 5000 MCP-1 receptors/cell. Differentiation of the cells induced by phorbol myristate acetate resulted in a 75% reduction of receptor gene expression within 2 h. Macrophage colony-stimulating factor had only moderate effect on hCCR2 expression. However, interferon γ inhibited MCP-1 binding by 60% at 48 h. The combination of macrophage colony-stimulating factor and interferon γ increased the inhibition to 80% at 48 h. This treatment has been shown previously to induce secretion of tumor necrosis factor α (TNF-α) and interleukin 1 (IL-1) in monocytes. Incubation of THP-1 cells with TNF-α and IL-1 induced a rapid down-regulation of hCCR2 expression and eventual loss of receptor protein. These cytokines exerted their regulatory role at the level of gene transcription. The effect of TNF-α alone persisted for 48 h, whereas the cells treated with IL-1 alone regained all of their receptor activity by 48 h. Our results suggest that cytokines can profoundly affect the expression of hCCR2 and thus modulate the recruitment of monocytes into sites of acute and chronic inflammation, including the developing atherosclerotic lesion.


Journal of Biological Chemistry | 2002

Regulation of Cholesterol Homeostasis and Lipid Metabolism in Skeletal Muscle by Liver X Receptors

George E. O. Muscat; Brandee L. Wagner; Jinzhao Hou; Rajendra K. Tangirala; Eric D. Bischoff; Paul R. Rohde; Mary Petrowski; Jiali Li; Gang Shao; Griffin Macondray; Ira G. Schulman

Recent studies have identified the liver X receptors (LXRα and LXRβ) as important regulators of cholesterol and lipid metabolism. Although originally identified as liver-enriched transcription factors, LXRs are also expressed in skeletal muscle, a tissue that accounts for ∼40% of human total body weight and is the major site of glucose utilization and fatty acid oxidation. Nevertheless, no studies have yet addressed the functional role of LXRs in muscle. In this work we utilize a combination of in vivoand in vitro analysis to demonstrate that LXRs can functionally regulate genes involved in cholesterol metabolism in skeletal muscle. Furthermore we show that treatment of muscle cellsin vitro with synthetic agonists of LXR increases the efflux of intracellular cholesterol to extracellular acceptors such as high density lipoprotein, thus identifying this tissue as a potential important regulator of reverse cholesterol transport and high density lipoprotein levels. Additionally we demonstrate that LXRα and a subset of LXR target genes are induced during myogenesis, suggesting a role for LXR-dependent signaling in the differentiation process.

Collaboration


Dive into the Rajendra K. Tangirala's collaboration.

Top Co-Authors

Avatar

Jason Kim

University of California

View shared research outputs
Top Co-Authors

Avatar

Peter Tontonoz

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cynthia Hong

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Grant D. Barish

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge