Network


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

Hotspot


Dive into the research topics where Jennifer H. Madenspacher is active.

Publication


Featured researches published by Jennifer H. Madenspacher.


Journal of Immunology | 2008

Effects of Liver X Receptor Agonist Treatment on Pulmonary Inflammation and Host Defense

Kathleen Smoak; Jennifer H. Madenspacher; Samithamby Jeyaseelan; Belinda Williams; Darlene Dixon; Katie R. Poch; Jerry A. Nick; G. Scott Worthen; Michael B. Fessler

Liver X receptor (LXR) α and β are members of the nuclear receptor superfamily of ligand-activated transcription factors. Best known for triggering “reverse cholesterol transport” gene programs upon their activation by endogenous oxysterols, LXRs have recently also been implicated in regulation of innate immunity. In this study, we define a role for LXRs in regulation of pulmonary inflammation and host defense and identify the lung and neutrophil as novel in vivo targets for pharmacologic LXR activation. LXR is expressed in murine alveolar macrophages, alveolar epithelial type II cells, and neutrophils. Treatment of mice with TO-901317, a synthetic LXR agonist, reduces influx of neutrophils to the lung triggered by inhaled LPS, intratracheal KC chemokine, and intratracheal Klebsiella pneumoniae and impairs pulmonary host defense against this bacterium. Pharmacologic LXR activation selectively modulates airspace cytokine expression induced by both LPS and K. pneumoniae. Moreover, we report for the first time that LXR activation impairs neutrophil motility and identify inhibition of chemokine-induced RhoA activation as a putative underlying mechanism. Taken together, these data define a novel role for LXR in lung pathophysiology and neutrophil biology and identify pharmacologic activation of LXR as a potential tool for modulation of innate immunity in the lung.


Journal of Pharmacology and Experimental Therapeutics | 2006

Alterations in the Rat Serum Proteome During Liver Injury from Acetaminophen Exposure

B. Alex Merrick; Maribel Bruno; Jennifer H. Madenspacher; Barbara A. Wetmore; Julie F. Foley; Rembert Pieper; Ming Zhao; Anthony J. Makusky; Andrew M. McGrath; Jeff Zhou; John L. Taylor; Kenneth B. Tomer

Changes in the serum proteome were identified during early, fulminant, and recovery phases of liver injury from acetaminophen in the rat. Male F344 rats received a single, noninjury dose or a high, injury-producing dose of acetaminophen for evaluation at 6 to 120 h. Two-dimensional gel electrophoresis of immunodepleted serum separated approximately 800 stained proteins per sample from which differentially expressed proteins were identified by mass spectrometry. Serum alanine aminotransferase/aspartate aminotransferase levels and histopathology revealed the greatest liver damage at 24 and 48 h after high-dose acetaminophen corresponding to the time of greatest serum protein alterations. After 24 h, 68 serum proteins were significantly altered of which 23 proteins were increased by >5-fold and 20 proteins were newly present compared with controls. Only minimal changes in serum proteins were noted at the low dose without any histopathology. Of the 54 total protein isoforms identified by mass spectrometry, gene ontology processes for 38 unique serum proteins revealed involvement of acute phase response, coagulation, protein degradation, intermediary metabolism, and various carrier proteins. Elevated serum tumor necrosis factor-α from 24 to 48 h suggested a mild inflammatory response accompanied by increased antioxidant capability demonstrated by increased serum catalase activity. Antibody array and enzyme-linked immunosorbent assay analyses also showed elevation in the chemokine monocyte chemoattractant protein-1 and the metalloprotease inhibitor tissue inhibitor of metalloproteinases-1 during this same period of liver injury. This study demonstrates that serum proteome alterations probably reflect both liver damage and a concerted, complex response of the body for organ repair and recovery during acute hepatic injury.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Combined Therapy of Dietary Fish Oil and Stearoyl-CoA Desaturase 1 Inhibition Prevents the Metabolic Syndrome and Atherosclerosis

J. Mark Brown; Soonkyu Chung; Janet K. Sawyer; Chiara Degirolamo; Heather M. Alger; Tam Nguyen; Xuewei Zhu; My Ngan Duong; Amanda L. Brown; Caleb C. Lord; Ramesh Shah; Matthew A. Davis; Kathryn Kelley; Martha D. Wilson; Jennifer H. Madenspacher; Michael B. Fessler; John S. Parks; Lawrence L. Rudel

Background—Stearoyl-CoA desaturase 1 (SCD1) is a critical regulator of energy metabolism and inflammation. We have previously reported that inhibition of SCD1 in hyperlipidemic mice fed a saturated fatty acid (SFA)-enriched diet prevented development of the metabolic syndrome, yet surprisingly promoted severe atherosclerosis. In this study we tested whether dietary fish oil supplementation could prevent the accelerated atherosclerosis caused by SCD1 inhibition. Methods and Results—LDLr−/−, ApoB100/100 mice were fed diets enriched in saturated fat or fish oil in conjunction with antisense oligonucleotide (ASO) treatment to inhibit SCD1. As previously reported, in SFA-fed mice, SCD1 inhibition dramatically protected against development of the metabolic syndrome, yet promoted atherosclerosis. In contrast, in mice fed fish oil, SCD1 inhibition did not result in augmented macrophage inflammatory response or severe atherosclerosis. In fact, the combined therapy of dietary fish oil and SCD1 ASO treatment effectively prevented both the metabolic syndrome and atherosclerosis. Conclusions—SCD1 ASO treatment in conjunction with dietary fish oil supplementation is an effective combination therapy to comprehensively combat the metabolic syndrome and atherosclerosis in mice.


The Journal of Allergy and Clinical Immunology | 2009

Novel relationship of serum cholesterol with asthma and wheeze in the United States.

Michael B. Fessler; M. Massing; Brian Spruell; Renee Jaramillo; David W. Draper; Jennifer H. Madenspacher; Samuel J. Arbes; Agustin Calatroni; Darryl C. Zeldin

BACKGROUND Cholesterol exerts complex effects on inflammation. There has been little investigation of whether serum cholesterol is associated with asthma, an inflammatory airways disease with great public health impact. OBJECTIVE To determine relationships between levels of 3 serum cholesterol measures (total cholesterol [TC], high-density lipoprotein cholesterol [HDL-C], and non-HDL-C) and asthma/wheeze in a sample representative of the US population. METHODS Cross-sectional study of 7005 participants age >or=6 years from the 2005 to 2006 National Health and Nutrition Examination Survey. RESULTS Serum TC and non-HDL-C were lower in patients with current asthma than in subjects without current asthma in the overall population (TC, 188.5 vs 192.2 mg/dL; non-HDL-C, 133.9 vs 137.7 mg/dL; P < .05 for both), whereas HDL-C was not different. Adjusted odds ratios (ORs) from multivariate logistic regression per 1-SD increase of TC and non-HDL-C for current asthma were 0.92 (95% CI, 0.86-0.98) and 0.91 (95% CI, 0.85-0.98), respectively. On racial/ethnic stratification, these relationships reflect marked reductions unique to Mexican Americans (MAs; TC, 171.4 vs 189.3 mg/dL; P < .001; OR, 0.62; 95% CI, 0.48-0.80; non-HDL-C, 119.8 vs 137.9 mg/dL; P < .001; OR, 0.62; 95% CI, 0.48-0.79). Among MAs, the adjusted OR for wheeze requiring medical attention was 0.57 (95% CI, 0.43-0.75) for TC and 0.53 (95% CI, 0.33-0.85) for non-HDL-C. Relationships between cholesterol and asthma/wheeze were independent of body mass index and serum C-reactive protein, and similar between atopic and nonatopic participants. CONCLUSION Serum TC and non-HDL-C are inversely related to asthma in the US population, chiefly reflecting a relationship among MAs.


Diabetes | 2012

CGI-58/ABHD5-Derived Signaling Lipids Regulate Systemic Inflammation and Insulin Action

Caleb C. Lord; Jenna L. Betters; Pavlina T. Ivanova; Stephen B. Milne; David S. Myers; Jennifer H. Madenspacher; Gwynneth Thomas; Soonkyu Chung; Mingxia Liu; Matthew A. Davis; Richard G. Lee; Rosanne M. Crooke; Mark J. Graham; John S. Parks; Dawn L. Brasaemle; Michael B. Fessler; H. Alex Brown; J. Mark Brown

Mutations of comparative gene identification 58 (CGI-58) in humans cause Chanarin-Dorfman syndrome, a rare autosomal recessive disease in which excess triacylglycerol (TAG) accumulates in multiple tissues. CGI-58 recently has been ascribed two distinct biochemical activities, including coactivation of adipose triglyceride lipase and acylation of lysophosphatidic acid (LPA). It is noteworthy that both the substrate (LPA) and the product (phosphatidic acid) of the LPA acyltransferase reaction are well-known signaling lipids. Therefore, we hypothesized that CGI-58 is involved in generating lipid mediators that regulate TAG metabolism and insulin sensitivity. Here, we show that CGI-58 is required for the generation of signaling lipids in response to inflammatory stimuli and that lipid second messengers generated by CGI-58 play a critical role in maintaining the balance between inflammation and insulin action. Furthermore, we show that CGI-58 is necessary for maximal TH1 cytokine signaling in the liver. This novel role for CGI-58 in cytokine signaling may explain why diminished CGI-58 expression causes severe hepatic lipid accumulation yet paradoxically improves hepatic insulin action. Collectively, these findings establish that CGI-58 provides a novel source of signaling lipids. These findings contribute insight into the basic mechanisms linking TH1 cytokine signaling to nutrient metabolism.


Journal of Immunology | 2010

Dyslipidemia Induces Opposing Effects on Intrapulmonary and Extrapulmonary Host Defense through Divergent TLR Response Phenotypes

Jennifer H. Madenspacher; David W. Draper; Kathleen Smoak; Haitao Li; Gary L. Griffiths; Benjamin T. Suratt; Martha D. Wilson; Lawrence L. Rudel; Michael B. Fessler

Dyslipidemia influences innate immune responses in the bloodstream, but whether and how pulmonary innate immunity is sensitive to circulating lipoproteins is largely unknown. To define whether dyslipidemia impacts responses to bacteria in the airspace and, if so, whether differently from its effects in other tissues, airspace, bloodstream, and i.p. responses to LPS and Klebsiella pneumoniae were investigated using murine models of dyslipidemia. Dyslipidemia reduced neutrophil (PMN) recruitment to the airspace in response to LPS and K. pneumoniae by impairing both chemokine induction in the airspace and PMN chemotaxis, thereby compromising pulmonary bacterial clearance. Paradoxically, bacteria were cleared more effectively from the bloodstream during dyslipidemia. This enhanced systemic response was due, at least in part, to basal circulating neutrophilia and basal TLR4/MyD88-dependent serum cytokine induction and enhanced serum cytokine responses to systemically administered TLR ligands. Dyslipidemia did not globally impair PMN transvascular trafficking to, and host defense within all loci, because neutrophilia, cytokine induction, and bacterial clearance were enhanced within the infected peritoneum. Peritoneal macrophages from dyslipidemic animals were primed for more robust TLR responses, reflecting increased lipid rafts and increased TLR4 expression, whereas macrophages from the airspace, in which cholesterol was maintained constant during dyslipidemia, had normal responses and rafts. Dyslipidemia thus imparts opposing effects upon intra- and extrapulmonary host defense by inducing tissue-divergent TLR response phenotypes and dysregulating airspace/blood compartmental levels of PMNs and cytokines. We propose that the airspace is a “privileged” site, thereby uniquely sensitive to dyslipidemia.


Cell Metabolism | 2010

Myeloid Differentiation Primary Response Protein 88 Couples Reverse Cholesterol Transport to Inflammation

Kathleen Smoak; Jim J. Aloor; Jennifer H. Madenspacher; B. Alex Merrick; Jennifer B. Collins; Xuewei Zhu; Giorgio Cavigiolio; Michael N. Oda; John S. Parks; Michael B. Fessler

Crosstalk exists in mammalian cells between cholesterol trafficking and innate immune signaling. Apolipoprotein A-I (apoA-I), a serum apolipoprotein that induces antiatherogenic efflux of macrophage cholesterol, is widely described as anti-inflammatory because it neutralizes bacterial lipopolysaccharide. Conversely, lipopolysaccharide-induced inflammation is proatherogenic. However, whether innate immunity plays an endogenous, physiological role in host cholesterol homeostasis in the absence of infection is undetermined. We report that apoA-I signals in the macrophage through Toll-like receptor (TLR)2, TLR4, and CD14, utilizing myeloid differentiation primary response protein 88 (MyD88)-dependent and -independent pathways, to activate nuclear factor-kappaB and induce cytokines. MyD88 plays a critical role in reverse cholesterol transport in vitro and in vivo, in part through promoting ATP-binding cassette A1 transporter upregulation. Taken together, this work identifies apoA-I as an endogenous stimulus of innate immunity that couples cholesterol trafficking to inflammation through MyD88 and identifies innate immunity as a physiologic signal in cholesterol homeostasis.


Journal of Experimental Medicine | 2013

p53 Integrates host defense and cell fate during bacterial pneumonia.

Jennifer H. Madenspacher; Kathleen M. Azzam; Kymberly M. Gowdy; Kenneth C. Malcolm; Jerry A. Nick; Darlene Dixon; Jim J. Aloor; David W. Draper; John Guardiola; Maria Shatz; Daniel Menendez; Julie Lowe; Jun Lu; Pierre R. Bushel; Leping Li; B. Alex Merrick; Michael A. Resnick; Michael B. Fessler

p53 deletion augments neutrophil-mediated bacterial clearance in the lung at the expense of tissue homeostasis, leading to increased mortality.


Journal of Cell Biology | 2013

p53 integrates host defense and cell fate during bacterial pneumonia

Jennifer H. Madenspacher; Kathleen M. Azzam; Kymberly M. Gowdy; Kenneth C. Malcolm; Jerry A. Nick; Darlene Dixon; Jim J. Aloor; David W. Draper; John Guardiola; Maria Shatz; Daniel Menendez; Julie Lowe; Jun Lu; Pierre R. Bushel; Leping Li; B. Alex Merrick; Michael A. Resnick; Michael B. Fessler

Cancer and infection are predominant causes of human mortality and derive, respectively, from inadequate genomic and host defenses against environmental agents. The transcription factor p53 plays a central role in human tumor suppression. Despite its expression in immune cells and broad responsiveness to stressors, it is virtually unknown whether p53 regulates host defense against infection. We report that the lungs of naive p53 / mice display genome-wide induction of NF-B response element–enriched proinflammatory genes, suggestive of type 1 immune priming. p53-null and p53 inhibitor–treated mice clear Gram-negative and -positive bacteria more effectively than controls after intrapulmonary infection. This is caused, at least in part, by cytokines produced by an expanded population of apoptosis-resistant, TLR-hyperresponsive alveolar macrophages that enhance airway neutrophilia. p53 / neutrophils, in turn, display heightened phagocytosis, Nox-dependent


Journal of Immunology | 2012

ATP Binding Cassette Transporter G1 Deletion Induces IL-17–Dependent Dysregulation of Pulmonary Adaptive Immunity

David W. Draper; Kymberly M. Gowdy; Jennifer H. Madenspacher; Rhonda H. Wilson; Gregory S. Whitehead; Hideki Nakano; Arun R. Pandiri; Julie F. Foley; Alan T. Remaley; Donald N. Cook; Michael B. Fessler

Mice with genetic deletion of the cholesterol transporter ATP binding cassette G1 (ABCG1) have pulmonary lipidosis and enhanced innate immune responses in the airway. Whether ABCG1 regulates adaptive immune responses to the environment is unknown. To this end, Abcg1+/+ and Abcg1−/− mice were sensitized to OVA via the airway using low-dose LPS as an adjuvant, and then challenged with OVA aerosol. Naive Abcg1−/− mice displayed increased B cells, CD4+ T cells, CD8+ T cells, and dendritic cells (DCs) in lung and lung-draining mediastinal lymph nodes, with lung CD11b+ DCs displaying increased CD80 and CD86. Upon allergen sensitization and challenge, the Abcg1−/− airway, compared with Abcg1+/+, displayed reduced Th2 responses (IL-4, IL-5, eosinophils), increased neutrophils and IL-17, but equivalent airway hyperresponsiveness. Reduced Th2 responses were also found using standard i.p. OVA sensitization with aluminum hydroxide adjuvant. Mediastinal lymph nodes from airway-sensitized Abcg1−/− mice produced reduced IL-5 upon ex vivo OVA challenge. Abcg1−/− CD4+ T cells displayed normal ex vivo differentiation, whereas Abcg1−/− DCs were found paradoxically to promote Th2 polarization. Th17 cells, IL-17+ γδT cells, and IL-17+ neutrophils were all increased in Abcg1−/− lungs, suggesting Th17 and non-Th17 sources of IL-17 excess. Neutralization of IL-17 prior to challenge normalized eosinophils and reduced neutrophilia in the Abcg1−/− airway. We conclude that Abcg1−/− mice display IL-17–mediated suppression of eosinophilia and enhancement of neutrophilia in the airway following allergen sensitization and challenge. These findings identify ABCG1 as a novel integrator of cholesterol homeostasis and adaptive immune programs.

Collaboration


Dive into the Jennifer H. Madenspacher's collaboration.

Top Co-Authors

Avatar

Michael B. Fessler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David W. Draper

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kymberly M. Gowdy

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jim J. Aloor

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kathleen Smoak

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

B. Alex Merrick

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alan T. Remaley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Darlene Dixon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jerry A. Nick

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge