Network


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

Hotspot


Dive into the research topics where Michelle N. Bradley is active.

Publication


Featured researches published by Michelle N. Bradley.


Cell | 2004

LXR-Dependent Gene Expression Is Important for Macrophage Survival and the Innate Immune Response

Sean B. Joseph; Michelle N. Bradley; Antonio Castrillo; Kevin W. Bruhn; Puiying A. Mak; Liming Pei; John B. Hogenesch; Ryan M. O'Connell; Genhong Cheng; Enrique Saez; Jeffery F. Miller; Peter Tontonoz

The liver X receptors (LXRs) are nuclear receptors with established roles in the regulation of lipid metabolism. We now show that LXR signaling not only regulates macrophage cholesterol metabolism but also impacts antimicrobial responses. Mice lacking LXRs are highly susceptible to infection with the intracellular bacteria Listeria monocytogenes (LM). Bone marrow transplant studies point to altered macrophage function as the major determinant of susceptibility. LXR-null macrophages undergo accelerated apoptosis when challenged with LM and exhibit defective bacterial clearance in vivo. These defects result, at least in part, from loss of regulation of the antiapoptotic factor SPalpha, a direct target for regulation by LXRalpha. Expression of LXRalpha or SPalpha in macrophages inhibits apoptosis in the setting of LM infection. Our results demonstrate that LXR-dependent gene expression plays an unexpected role in innate immunity and suggest that common nuclear receptor pathways mediate macrophage responses to modified lipoproteins and intracellular pathogens.


Cell | 2008

LXR signaling couples sterol metabolism to proliferation in the acquired immune response

Steven J. Bensinger; Michelle N. Bradley; Sean B. Joseph; Noam Zelcer; Edith M. Janssen; Mary Ann Hausner; Roger Shih; John S. Parks; Peter A. Edwards; Beth D. Jamieson; Peter Tontonoz

Cholesterol is essential for membrane synthesis; however, the mechanisms that link cellular lipid metabolism to proliferation are incompletely understood. We demonstrate here that cellular cholesterol levels in dividing T cells are maintained in part through reciprocal regulation of the LXR and SREBP transcriptional programs. T cell activation triggers induction of the oxysterol-metabolizing enzyme SULT2B1, consequent suppression of the LXR pathway for cholesterol transport, and promotion of the SREBP pathway for cholesterol synthesis. Ligation of LXR during T cell activation inhibits mitogen-driven expansion, whereas loss of LXRbeta confers a proliferative advantage. Inactivation of the sterol transporter ABCG1 uncouples LXR signaling from proliferation, directly linking sterol homeostasis to the antiproliferative action of LXR. Mice lacking LXRbeta exhibit lymphoid hyperplasia and enhanced responses to antigenic challenge, indicating that proper regulation of LXR-dependent sterol metabolism is important for immune responses. These results implicate LXR signaling in a metabolic checkpoint that modulates cell proliferation and immunity.


Immunity | 2009

Apoptotic Cells Promote Their Own Clearance and Immune Tolerance through Activation of the Nuclear Receptor LXR

Noelia A-Gonzalez; Steven J. Bensinger; Cynthia Hong; Susana Beceiro; Michelle N. Bradley; Noam Zelcer; José Manuel Deniz; Cristina M. Ramírez; Mercedes Diaz; Germán Gallardo; Carlos M. Ruiz de Galarreta; Jon Salazar; Felix Lopez; Peter A. Edwards; John S. Parks; Miguel Andujar; Peter Tontonoz; Antonio Castrillo

Effective clearance of apoptotic cells by macrophages is essential for immune homeostasis. The transcriptional pathways that allow macrophages to sense and respond to apoptotic cells are poorly defined. We found that liver X receptor (LXR) signaling was important for both apoptotic cell clearance and the maintenance of immune tolerance. Apoptotic cell engulfment activated LXR and thereby induced the expression of Mer, a receptor tyrosine kinase critical for phagocytosis. LXR-deficient macrophages exhibited a selective defect in phagocytosis of apoptotic cells and an aberrant proinflammatory response to them. As a consequence of these defects, mice lacking LXRs manifested a breakdown in self-tolerance and developed autoantibodies and autoimmune glomerulonephritis. Treatment with an LXR agonist ameliorated disease progression in a mouse model of lupus-like autoimmunity. Thus, activation of LXR by apoptotic cells engages a virtuous cycle that promotes their own clearance and couples engulfment to the suppression of inflammatory pathways.


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.


Nature Immunology | 2001

Nucleosome remodeling at the IL-12 p40 promoter is a TLR-dependent, Rel-independent event.

Amy S. Weinmann; Deborah M. Mitchell; Shomyseh Sanjabi; Michelle N. Bradley; Alexander Hoffmann; Hsiou Chi Liou; Stephen T. Smale

Lipopolysaccharide (LPS) induction of the gene encoding interleukin 12 p40 requires remodeling of a promoter-encompassing nucleosome and the Toll-like receptor (TLR)–mediated activation of a c-Rel–containing complex. Analysis of TLR4-mutant mice revealed that remodeling requires TLR signaling. However, Rel proteins and other proteins required for transcription of an integrated p40 promoter were insufficient for remodeling. c-Rel was also unnecessary for remodeling, as remodeling was observed in c-Rel−/− macrophages, which lack p40 transcripts. These results suggest that remodeling requires TLR signaling pathways that diverge from the c-Rel activation pathways. The factors that stimulate remodeling may represent, therefore, newly identified targets of TLR signaling and of agents that regulate inflammatory responses and TH1 development.


Journal of Clinical Investigation | 2010

LXR promotes the maximal egress of monocyte-derived cells from mouse aortic plaques during atherosclerosis regression

Jonathan E. Feig; Inés Pineda-Torra; Marie Sanson; Michelle N. Bradley; Yuliya Vengrenyuk; Dusan Bogunovic; Emmanuel L. Gautier; Daniel Rubinstein; Cynthia Hong; Jianhua Liu; Chaowei Wu; Nico van Rooijen; Nina Bhardwaj; Michael J. Garabedian; Peter Tontonoz; Edward A. Fisher

We have previously shown that mouse atherosclerosis regression involves monocyte-derived (CD68+) cell emigration from plaques and is dependent on the chemokine receptor CCR7. Concurrent with regression, mRNA levels of the gene encoding LXRalpha are increased in plaque CD68+ cells, suggestive of a functional relationship between LXR and CCR7. To extend these results, atherosclerotic Apoe-/- mice sufficient or deficient in CCR7 were treated with an LXR agonist, resulting in a CCR7-dependent decrease in plaque CD68+ cells. To test the requirement for LXR for CCR7-dependent regression, we transplanted aortic arches from atherosclerotic Apoe-/- mice, or from Apoe-/- mice with BM deficiency of LXRalpha or LXRbeta, into WT recipients. Plaques from both LXRalpha and LXRbeta-deficient Apoe-/- mice exhibited impaired regression. In addition, the CD68+ cells displayed reduced emigration and CCR7 expression. Using an immature DC line, we found that LXR agonist treatment increased Ccr7 mRNA levels. This increase was blunted when LXRalpha and LXRbeta levels were reduced by siRNAs. Moreover, LXR agonist treatment of primary human immature DCs resulted in functionally significant upregulation of CCR7. We conclude that LXR is required for maximal effects on plaque CD68+ cell expression of CCR7 and monocyte-derived cell egress during atherosclerosis regression in mice.


Molecular and Cellular Biology | 2003

C/EBPβ Regulation in Lipopolysaccharide-Stimulated Macrophages

Michelle N. Bradley; Liang Zhou; Stephen T. Smale

ABSTRACT C/EBP family members contribute to the induction of the interleukin-12 p40 gene and the genes encoding several other mediators of inflammation. Here, we show by chromatin immunoprecipitation that C/EBPβ binds the p40 promoter following lipopolysaccharide stimulation of peritoneal macrophages. However, three modes of C/EBPβ regulation reported in other cell types were not detected, including alternative translation initiation, nuclear translocation, and increased DNA binding following posttranslational modification. In contrast, C/EBPβ concentrations greatly increased following stimulation via MAP kinase-dependent induction of C/EBPβ gene transcription. Increased C/EBPβ concentrations were unimportant for p40 induction, however, as transcription of the p40 gene initiated before C/EBPβ concentrations increased. Furthermore, disruption of C/EBPβ upregulation by a MAP kinase inhibitor only slightly diminished p40 induction. Phosphopeptide mapping revealed that endogenous C/EBPβ in macrophages is phosphorylated on only a single tryptic peptide containing 14 potential phosphoacceptors. This peptide was constitutively phosphorylated in primary and transformed macrophages, in contrast to its inducible phosphorylation in other cell types in response to Ras and growth hormone signaling. Altered-specificity experiments supported the hypothesis that C/EBPβ activity in macrophages does not require an inducible posttranslational modification. These findings suggest that, although C/EBPβ contributes to the induction of numerous proinflammatory genes, it is fully active in unstimulated macrophages and poised to stimulate transcription in conjunction with other factors whose activities are induced.


Journal of Immunology | 2008

TLR/MyD88 and liver X receptor alpha signaling pathways reciprocally control Chlamydia pneumoniae-induced acceleration of atherosclerosis.

Yoshikazu Naiki; Rosalinda Sorrentino; Michelle H. Wong; Kathrin S. Michelsen; Kenichi Shimada; Shuang Chen; Atilla Yilmaz; Anatoly Slepenkin; Nicolas W. J. Schröder; Timothy R. Crother; Yonca Bulut; Terence M. Doherty; Michelle N. Bradley; Zory Shaposhnik; Ellena M. Peterson; Peter Tontonoz; Prediman K. Shah; Moshe Arditi

Experimental and clinical studies link Chlamydia pneumoniae infection to atherogenesis and atherothrombotic events, but the underlying mechanisms are unclear. We tested the hypothesis that C. pneumoniae-induced acceleration of atherosclerosis in apolipoprotein E (ApoE)−/− mice is reciprocally modulated by activation of TLR-mediated innate immune and liver X receptor α (LXRα) signaling pathways. We infected ApoE−/− mice and ApoE−/− mice that also lacked TLR2, TLR4, MyD88, or LXRα intranasally with C. pneumoniae followed by feeding of a high fat diet for 4 mo. Mock-infected littermates served as controls. Atherosclerosis was assessed in aortic sinuses and in en face preparation of whole aorta. The numbers of activated dendritic cells (DCs) within plaques and the serum levels of cholesterol and proinflammatory cytokines were also measured. C. pneumoniae infection markedly accelerated atherosclerosis in ApoE-deficient mice that was associated with increased numbers of activated DCs in aortic sinus plaques and higher circulating levels of MCP-1, IL-12p40, IL-6, and TNF-α. In contrast, C. pneumoniae infection had only a minimal effect on atherosclerosis, accumulation of activated DCs in the sinus plaques, or circulating cytokine increases in ApoE−/− mice that were also deficient in TLR2, TLR4, or MyD88. However, C. pneumoniae-induced acceleration of atherosclerosis in ApoE−/− mice was further enhanced in ApoE−/−LXRα−/− double knockout mice and was accompanied by higher serum levels of IL-6 and TNF-α. We conclude that C. pneumoniae infection accelerates atherosclerosis in hypercholesterolemic mice predominantly through a TLR/MyD88-dependent mechanism and that LXRα appears to reciprocally modulate and reduce the proatherogenic effects of C. pneumoniae infection.


Journal of Biological Chemistry | 2006

The Arginase II Gene Is an Anti-inflammatory Target of Liver X Receptor in Macrophages

Chaitra Marathe; Michelle N. Bradley; Cynthia Hong; Felix Lopez; Carlos M. Ruiz de Galarreta; Peter Tontonoz; Antonio Castrillo

The liver X receptors (LXRs) are ligand-dependent transcription factors that have been implicated in lipid metabolism and inflammation. LXRs also inhibit the expression of inflammatory genes in macrophages, including inducible nitric oxide synthase (iNOS). Some of these actions are mediated through LXR antagonism of NF-κB activity. The potential for LXRs to positively regulate the expression of anti-inflammatory molecules, however, has not been explored. Here we show that the arginase II (ArgII) gene is a direct target for LXR regulation. ArgII catalyzes the conversion of l-arginine into l-ornithine and urea, leading to the synthesis of polyamines. Expression of ArgII is induced by LXR agonists in macrophage cell lines and primary murine macrophages in a receptor-dependent manner. The ArgII promoter contains a functional LXR response elements that mediates promoter induction by LXR/RXR (retinoid X receptor) in transfection assays. Since ArgII and iNOS utilize a common substrate, induction of ArgII expression has the potential to exert anti-inflammatory effects by shifting arginine metabolism toward polyamine synthesis at the expense of NO production. In support of this hypothesis, we demonstrate that forced expression of ArgII mimics the inhibitory effect of LXR activation on macrophage NO production. Furthermore, inhibition of arginase activity partially reverses the inhibitory effect of LXR agonists on NO production. These studies suggest that regulation of ArgII may contribute to the immunomodulatory effects of LXRs.


Journal of Medicinal Chemistry | 2010

The Identification of Indacaterol as an Ultralong-Acting Inhaled β2-Adrenoceptor Agonist

François Baur; David Beattie; David Beer; David Bentley; Michelle N. Bradley; Ian Bruce; Steven J. Charlton; Bernard Cuenoud; Roland Ernst; Robin Alec Fairhurst; Bernard Faller; David Farr; Thomas H. Keller; John R. Fozard; Joe Fullerton; Sheila Garman; Julia Hatto; Claire Hayden; Handan He; Colin Howes; Diana Janus; Zhengjin Jiang; Christine Lewis; Frédérique Loeuillet-Ritzler; Heinz E. Moser; John Reilly; Alan Steward; David A. Sykes; Lauren Tedaldi; Alexandre Trifilieff

Following a lipophilicity-based hypothesis, an 8-hydroxyquinolinone 2-aminoindan derived series of beta(2)-adrenoceptor agonists have been prepared and evaluated for their potential as inhaled ultralong-acting bronchodilators. Determination of their activities at the human beta(2)-adrenoceptor receptor showed symmetrical substitution of the 2-aminoindan moiety at the 5- and 6-positions delivered the targeted intermediate potency and intrinsic-efficacy profiles relative to a series of clinical reference beta(2)-adrenoceptor agonists. Further assessment with an in vitro superfused electrically stimulated guinea-pig tracheal-strip assay established the onset and duration of action time courses, which could be rationalized by considering the lipophilicity, potency, and intrinsic efficacy of the compounds. From these studies the 5,6-diethylindan analogue indacaterol 1c was shown to possess a unique profile of combining a rapid onset of action with a long duration of action. Further in vivo profiling of 1c supported the long duration of action and a wide therapeutic index following administration to the lung, which led to the compound being selected as a development candidate.

Collaboration


Dive into the Michelle N. Bradley's collaboration.

Top Co-Authors

Avatar

Peter Tontonoz

University of California

View shared research outputs
Top Co-Authors

Avatar

Cynthia Hong

University of California

View shared research outputs
Top Co-Authors

Avatar

Jon Salazar

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio Castrillo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chaitra Marathe

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge