Network


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

Hotspot


Dive into the research topics where Bryan A. Laffitte is active.

Publication


Featured researches published by Bryan A. Laffitte.


Molecular Cell | 2001

A PPARγ-LXR-ABCA1 Pathway in Macrophages Is Involved in Cholesterol Efflux and Atherogenesis

Ajay Chawla; William A. Boisvert; Chih-Hao Lee; Bryan A. Laffitte; Yaacov Barak; Sean B. Joseph; Debbie Liao; Laszlo Nagy; Peter A. Edwards; Linda K. Curtiss; Ronald M. Evans; Peter Tontonoz

Abstract Previous work has implicated PPARγ in the regulation of CD36 expression and macrophage uptake of oxidized LDL (oxLDL). We provide evidence here that in addition to lipid uptake, PPARγ regulates a pathway of cholesterol efflux. PPARγ induces ABCA1 expression and cholesterol removal from macrophages through a transcriptional cascade mediated by the nuclear receptor LXRα. Ligand activation of PPARγ leads to primary induction of LXRα and to coupled induction of ABCA1. Transplantation of PPARγ null bone marrow into LDLR −/− mice results in a significant increase in atherosclerosis, consistent with the hypothesis that regulation of LXRα and ABCA1 expression is protective in vivo. Thus, we propose that PPARγ coordinates a complex physiologic response to oxLDL that involves particle uptake, processing, and cholesterol removal through ABCA1.


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

Synthetic LXR ligand inhibits the development of atherosclerosis in mice

Sean B. Joseph; Elaine McKilligin; Liming Pei; Michael A. Watson; Alan R. Collins; Bryan A. Laffitte; Mingyi Chen; Grace Noh; Joanne Goodman; Graham N. Hagger; Jonathan Tran; Tim K. Tippin; Xuping Wang; Aldons J. Lusis; Willa A. Hsueh; Ronald E. Law; Jon L. Collins; Timothy M. Willson; Peter Tontonoz

The nuclear receptors LXRα and LXRβ have been implicated in the control of cholesterol and fatty acid metabolism in multiple cell types. Activation of these receptors stimulates cholesterol efflux in macrophages, promotes bile acid synthesis in liver, and inhibits intestinal cholesterol absorption, actions that would collectively be expected to reduce atherosclerotic risk. However, synthetic LXR ligands have also been shown to induce lipogenesis and hypertriglyceridemia in mice, raising questions as to the net effects of these compounds on the development of cardiovascular disease. We demonstrate here that the nonsteroidal LXR agonist GW3965 has potent antiatherogenic activity in two different murine models. In LDLR−/− mice, GW3965 reduced lesion area by 53% in males and 34% in females. A similar reduction of 47% was observed in male apoE−/− mice. Long-term (12-week) treatment with LXR agonist had differential effects on plasma lipid profiles in LDLR−/− and apoE−/− mice. GW3965 induced expression of ATP-binding cassettes A1 and G1 in modified low-density lipoprotein-loaded macrophages in vitro as well as in the aortas of hyperlipidemic mice, suggesting that direct actions of LXR ligands on vascular gene expression are likely to contribute to their antiatherogenic effects. These observations provide direct evidence for an atheroprotective effect of LXR agonists and support their further evaluation as potential modulators of human cardiovascular disease.


Journal of Biological Chemistry | 2002

Direct and Indirect Mechanisms for Regulation of Fatty Acid Synthase Gene Expression by Liver X Receptors

Sean B. Joseph; Bryan A. Laffitte; Parthive H. Patel; Michael A. Watson; Karen E. Matsukuma; Robert Walczak; Jon L. Collins; Timothy F. Osborne; Peter Tontonoz

The nuclear receptors LXRα and LXRβ have been implicated in the control of lipogenesis and cholesterol homeostasis. Ligand activation of these receptors in vivoinduces expression of the LXR target gene SREBP-1cand increases plasma triglyceride levels. Expression of fatty acid synthase (FAS), a central enzyme in de novo lipogenesis and an established target of the SREBP-1 pathway, is also induced by LXR ligands. The effects of LXR ligands on FAS expression have been proposed to be entirely secondary to the induction of SREBP-1c. We demonstrate here that LXRs regulate FAS expression through direct interaction with the FAS promoter as well as through activation of SREBP-1c expression. Induction of FAS expression in HepG2 cells by LXR ligands is reduced, but not abolished, under conditions where SREBP processing is suppressed. Moreover, LXR ligands induce FAS expression in CHO-7 cells without altering expression of SREBP-1. We demonstrate that in addition to tandem SREBP sites, the FASpromoter contains a high affinity binding site for the LXR/RXR heterodimer that is conserved in diverse animal species including birds, rodents, and humans. The LXR and SREBP binding sites independently confer LXR responsiveness on the FASpromoter, and maximal induction requires both transcription factors. Transient elevation of plasma triglyceride levels in mice treated with a synthetic LXR agonist correlates with transient induction of hepatic FAS expression. These results indicate that the LXR signaling pathway modulates FAS expression through distinct but complementary mechanisms and suggest that the FAS gene may be a critical target in the control of lipogenesis by LXRs.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue

Bryan A. Laffitte; Lily C. Chao; Jing Li; Robert Walczak; Sarah Hummasti; Sean B. Joseph; Antonio Castrillo; Damien C. Wilpitz; David J. Mangelsdorf; Jon L. Collins; Enrique Saez; Peter Tontonoz

The control of lipid and glucose metabolism is closely linked. The nuclear receptors liver X receptor (LXR)α and LXRβ have been implicated in gene expression linked to lipid homeostasis; however, their role in glucose metabolism is not clear. We demonstrate here that the synthetic LXR agonist GW3965 improves glucose tolerance in a murine model of diet-induced obesity and insulin resistance. Analysis of gene expression in LXR agonist-treated mice reveals coordinate regulation of genes involved in glucose metabolism in liver and adipose tissue. In the liver, activation of LXR led to the suppression of the gluconeogenic program including down-regulation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1), phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase expression. Inhibition of gluconeogenic genes was accompanied by an induction in expression of glucokinase, which promotes hepatic glucose utilization. In adipose tissue, activation of LXR led to the transcriptional induction of the insulin-sensitive glucose transporter, GLUT4. We show that the GLUT4 promoter is a direct transcriptional target for the LXR/retinoid X receptor heterodimer and that the ability of LXR ligands to induce GLUT4 expression is abolished in LXR null cells and animals. Consistent with their effects on GLUT4 expression, LXR agonists promote glucose uptake in 3T3-L1 adipocytes in vitro. Thus, activation of LXR alters the expression of genes in liver and adipose tissue that collectively would be expected to limit hepatic glucose output and improve peripheral glucose uptake. These results outline a role for LXRs in the coordination of lipid and glucose metabolism.


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.


Molecular and Cellular Biology | 2001

Autoregulation of the human liver X receptor alpha promoter.

Bryan A. Laffitte; Sean B. Joseph; Robert Walczak; Liming Pei; Damien C. Wilpitz; Jon L. Collins; Peter Tontonoz

ABSTRACT Previous work has implicated the nuclear receptors liver X receptor α (LXRα) and LXRβ in the regulation of macrophage gene expression in response to oxidized lipids. Macrophage lipid loading leads to ligand activation of LXRs and to induction of a pathway for cholesterol efflux involving the LXR target genes ABCA1 andapoE. We demonstrate here that autoregulation of the LXRα gene is an important component of this lipid-inducible efflux pathway in human macrophages. Oxidized low-density lipoprotein, oxysterols, and synthetic LXR ligands induce expression of LXRα mRNA in human monocyte-derived macrophages and human macrophage cell lines but not in murine peritoneal macrophages or cell lines. This is in contrast to peroxisome proliferator-activated receptor γ (PPARγ)-specific ligands, which stimulate LXRα expression in both human and murine macrophages. We further demonstrate that LXR and PPARγ ligands cooperate to induce LXRα expression in human but not murine macrophages. Analysis of the human LXRα promoter led to the identification of multiple LXR response elements. Interestingly, the previously identified PPAR response element (PPRE) in the murine LXRα gene is not conserved in humans; however, a different PPRE is present in the human LXR 5′-flanking region. These results have implications for cholesterol metabolism in human macrophages and its potential to be regulated by synthetic LXR and/or PPARγ ligands. The ability of LXRα to regulate its own promoter is likely to be an integral part of the macrophage physiologic response to lipid loading.


Journal of Biological Chemistry | 2002

Regulated Expression of the Apolipoprotein E/C-I/C-IV/C-II Gene Cluster in Murine and Human Macrophages A CRITICAL ROLE FOR NUCLEAR LIVER X RECEPTORS α AND β

Puiying A. Mak; Bryan A. Laffitte; Catherine Desrumaux; Sean B. Joseph; Linda K. Curtiss; David J. Mangelsdorf; Peter Tontonoz; Peter A. Edwards

Lipid-loaded macrophage “foam cells” accumulate in the subendothelial space during the development of fatty streaks and atherosclerotic lesions. To better understand the consequences of such lipid loading, murine peritoneal macrophages were isolated and incubated with ligands for two nuclear receptors, liver X receptor (LXR) and retinoic acid receptor (RXR). Analysis of the expressed mRNAs using microarray technology led to the identification of four highly induced genes that encode apolipoproteins E, C-I, C-IV, and C-II. Northern blot analysis confirmed that the mRNA levels of these four genes were induced 2–14-fold in response to natural or synthetic ligands for LXR and/or RXR. The induction of all four mRNAs was greatly attenuated in peritoneal macrophages derived from LXRα/β null mice. The two LXR response elements located within the multienhancers ME.1 and ME.2 were shown to be essential for the induction of apoC-II promoter-reporter genes by ligands for LXR and/or RXR. Finally, immunohistochemical studies demonstrate that apoC-II protein co-localizes with macrophages within murine arterial lesions. Taken together, these studies demonstrate that activated LXR induces the expression of the apoE/C-I/C-IV/C-II gene cluster in both human and murine macrophages. These results suggest an alternative mechanism by which lipids are removed from macrophage foam cells.


Molecular and Cellular Biology | 2003

The phospholipid transfer protein gene is a liver X receptor target expressed by macrophages in atherosclerotic lesions

Bryan A. Laffitte; Sean B. Joseph; Mingyi Chen; Antonio Castrillo; Joyce J. Repa; Damien C. Wilpitz; David J. Mangelsdorf; Peter Tontonoz

ABSTRACT The liver X receptors (LXRs) are members of the nuclear receptor superfamily that are activated by oxysterols. In response to ligand binding, LXRs regulate a variety of genes involved in the catabolism, transport, and uptake of cholesterol and its metabolites. Here we demonstrate that LXRs also regulate plasma lipoprotein metabolism through control of the phospholipid transfer protein (PLTP) gene. LXR ligands induce the expression of PLTP in cultured HepG2 cells and mouse liver in vivo in a coordinate manner with known LXR target genes. Moreover, plasma phospholipid transfer activity is increased in mice treated with the synthetic LXR ligand GW3965. Unexpectedly, PLTP expression was also highly inducible by LXR in macrophages, a cell type not previously recognized to express this enzyme. The ability of synthetic and oxysterol ligands to regulate PLTP mRNA in macrophages and liver is lost in animals lacking both LXRα and LXRβ, confirming the critical role of these receptors. We further demonstrate that the PLTP promoter contains a high-affinity LXR response element that is bound by LXR/RXR heterodimers in vitro and is activated by LXR/RXR in transient-transfection studies. Finally, immunohistochemistry studies reveal that PLTP is highly expressed by macrophages within human atherosclerotic lesions, suggesting a potential role for this enzyme in lipid-loaded macrophages. These studies outline a novel pathway whereby LXR and its ligands may modulate lipoprotein metabolism.


Journal of Lipid Research | 2004

Liver X receptors are regulators of adipocyte gene expression but not differentiation identification of apoD as a direct target

Sarah Hummasti; Bryan A. Laffitte; Michael A. Watson; Cristin M. Galardi; Lily C. Chao; Lakshman Ramamurthy; John T. Moore; Peter Tontonoz

The liver X receptors α and β (LXRα and LXRβ) have been shown to play important roles in lipid homeostasis in liver and macrophages, however, their function in adipose tissue is not well defined. Both LXRs are highly expressed in fat, and the expression of LXRα increases during adipogenesis. Furthermore, LXRα expression is induced by peroxisome proliferator-activated receptor γ (PPARγ), the master regulator of fat cell differentiation. Here we investigate the role of LXRs in adipocyte differentiation and gene expression and their potential crosstalk with the PPARγ pathway. We demonstrate that LXR agonists have no significant effect on the differentiation of 3T3-F442A or 3T3-L1 preadipocytes in vitro and do not alter the expression of differentiation-linked PPARγ target genes in vivo. Moreover, retroviral expression of LXRα in NIH-3T3 cells does not alter the adipogenic potential of these cells and neither augments nor inhibits the action of PPARγ. However, transcriptional profiling studies reveal that LXRs are important regulators of adipocyte gene expression. We identify the multifunction lipid carrier protein apolipoprotein D and the lipogenic protein Spot 14 as LXR responsive genes both in vitro and in vivo. Thus, although LXRs do not influence adipocyte differentiation per se, these receptors are likely to play an important role in the modulation of lipid metabolism in adipocytes.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Screening the mammalian extracellular proteome for regulators of embryonic human stem cell pluripotency

Rodolfo Gonzalez; Lori L. Jennings; Mark W. Knuth; Anthony P. Orth; Heath E. Klock; Weija Ou; Julie Feuerhelm; Mitchell V. Hull; Eric Koesema; Yuping Wang; Jia Zhang; Chunlei Wu; Charles Y. Cho; Andrew I. Su; Serge Batalov; Hong Chen; Kristen Johnson; Bryan A. Laffitte; Deborah G. Nguyen; Evan Y. Snyder; Peter G. Schultz; Jennifer L. Harris; Scott A. Lesley

Approximately 3,500 mammalian genes are predicted to be secreted or single-pass transmembrane proteins. The function of the majority of these genes is still unknown, and a number of the encoded proteins might find use as new therapeutic agents themselves or as targets for small molecule or antibody drug development. To analyze the physiological activities of the extracellular proteome, we developed a large-scale, high-throughput protein expression, purification, and screening platform. For this study, the complete human extracellular proteome was analyzed and prioritized based on genome-wide disease association studies to select 529 initial target genes. These genes were cloned into three expression vectors as native sequences and as N-terminal and C-terminal Fc fusions to create an initial collection of 806 purified secreted proteins. To determine its utility, this library was screened in an OCT4-based cellular assay to identify regulators of human embryonic stem-cell self-renewal. We found that the pigment epithelium-derived factor can promote long-term pluripotent growth of human embryonic stem cells without bFGF or TGFβ/Activin/Nodal ligand supplementation. Our results further indicate that activation of the pigment epithelium-derived factor receptor-Erk1/2 signaling pathway by the pigment epithelium-derived factor is sufficient to maintain the self-renewal of pluripotent human embryonic stem cells. These experiments illustrate the potential for discovering novel biological functions by directly screening protein diversity in cell-based phenotypic or reporter assays.

Collaboration


Dive into the Bryan A. Laffitte's collaboration.

Top Co-Authors

Avatar

Peter Tontonoz

University of California

View shared research outputs
Top Co-Authors

Avatar

Sean B. Joseph

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David J. Mangelsdorf

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Walczak

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter G. Schultz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge