Network


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

Hotspot


Dive into the research topics where Amy P. Patterson is active.

Publication


Featured researches published by Amy P. Patterson.


Infection and Immunity | 2002

Lipopolysaccharide Down Regulates Both Scavenger Receptor B1 and ATP Binding Cassette Transporter A1 in RAW Cells

Irina N. Baranova; Tatyana G. Vishnyakova; Alexander V. Bocharov; Zhigang Chen; Alan T. Remaley; John A. Stonik; Thomas L. Eggerman; Amy P. Patterson

ABSTRACT Lipopolysaccharide (LPS) has recently been shown to facilitate macrophage foam cell formation and has been suggested to be a proatherogenic factor. The mechanism of LPS induced cholesterol accumulation, however, is unclear. In this report, using the macrophage-like RAW 264.7 cell line, we provide experimental evidence that LPSs proatherogenic effects may at least in part reflect altered cholesterol metabolism. Data presented demonstrate that in a dose-dependent manner, LPS is able to down regulate the mRNA expression of the two primary high-density lipoprotein (HDL) receptors, scavenger receptor B1 (SR-B1) and ATP binding cassette A1 (ABCA1), with a 50% inhibitory concentration of less than 0.2 ng/ml, as well as to decrease SR-B1 protein expression by 80%. We also found that LPS treatment resulted in a significant decrease (to 20% of the control level) of the specific 125I-HDL binding as well as in 50% inhibition of the HDL-mediated cholesterol efflux compared to untreated cells. In addition, we compared the potencies of various modified LPS preparations and demonstrated that the phosphorylated lipid A portion of LPS, which is highly conserved among gram-negative microorganisms, including Chlamydia, is primarily responsible for the effects of LPS on SR-B1 and ABCA1 expression. Inhibitors of NF-κB activation were observed to efficiently block the suppressive effect of LPS on SR-B1 and ABCA1, suggesting a mechanism involving NF-κB. These data indicate that the LPS effects on cholesterol metabolism may contribute to the proatherogenic properties of LPS.


The New England Journal of Medicine | 2013

Research as a Part of Public Health Emergency Response

Nicole Lurie; Teri A. Manolio; Amy P. Patterson; Francis S. Collins; Thomas R. Frieden

The authors review lessons learned from several recent public health emergencies and argue that we must conduct research during emergencies to improve our capacity to prevent illness and injury. They propose policies to facilitate timely research.


Journal of Biological Chemistry | 2010

CD36 Is a Novel Serum Amyloid A (SAA) Receptor Mediating SAA Binding and SAA-induced Signaling in Human and Rodent Cells

Irina N. Baranova; Alexander V. Bocharov; Tatyana G. Vishnyakova; Roger J. Kurlander; Zhigang Chen; Dong Fu; Irwin M. Arias; Gyorgy Csako; Amy P. Patterson; Thomas L. Eggerman

Serum amyloid A (SAA) is a major acute phase protein involved in multiple physiological and pathological processes. This study provides experimental evidence that CD36, a phagocyte class B scavenger receptor, functions as a novel SAA receptor mediating SAA proinflammatory activity. The uptake of Alexa Fluor® 488 SAA as well as of other well established CD36 ligands was increased 5–10-fold in HeLa cells stably transfected with CD36 when compared with mock-transfected cells. Unlike other apolipoproteins that bind to CD36, only SAA induced a 10–50-fold increase of interleukin-8 secretion in CD36-overexpressing HEK293 cells when compared with control cells. SAA-mediated effects were thermolabile, inhibitable by anti-SAA antibody, and also neutralized by association with high density lipoprotein but not by association with bovine serum albumin. SAA-induced cell activation was inhibited by a CD36 peptide based on the CD36 hexarelin-binding site but not by a peptide based on the thrombospondin-1-binding site. A pronounced reduction (up to 60–75%) of SAA-induced pro-inflammatory cytokine secretion was observed in cd36−/− rat macrophages and Kupffer cells when compared with wild type rat cells. The results of the MAPK phosphorylation assay as well as of the studies with NF-κB and MAPK inhibitors revealed that two MAPKs, JNK and to a lesser extent ERK1/2, primarily contribute to elevated cytokine production in CD36-overexpressing HEK293 cells. In macrophages, four signaling pathways involving NF-κB and three MAPKs all appeared to contribute to SAA-induced cytokine release. These observations indicate that CD36 is a receptor mediating SAA binding and SAA-induced pro-inflammatory cytokine secretion predominantly through JNK- and ERK1/2-mediated signaling.


Proceedings of the National Academy of Sciences of the United States of America | 2006

CLA-1 and its splicing variant CLA-2 mediate bacterial adhesion and cytosolic bacterial invasion in mammalian cells

Tatyana G. Vishnyakova; Roger Kurlander; Alexander V. Bocharov; Irina N. Baranova; Zhigang Chen; Mones Abu-Asab; Maria Tsokos; Daniela Malide; Federica Basso; Alan T. Remaley; Gyorgy Csako; Thomas L. Eggerman; Amy P. Patterson

CD36 and LIMPII analog 1, CLA-1, and its splicing variant, CLA-2 (SR-BI and SR-BII in rodents), are human high density lipoprotein receptors with an identical extracellular domain which binds a spectrum of ligands including bacterial cell wall components. In this study, CLA-1- and CLA-2-stably transfected HeLa and HEK293 cells demonstrated several-fold increases in the uptake of various bacteria over mock-transfected cells. All bacteria tested, including both Gram-negatives (Escherichia coli K12, K1 and Salmonella typhimurium) and Gram-positives (Staphylococcus aureus and Listeria monocytogenes), demonstrated various degrees of lower uptake in control cells. This result is consistent with the presence of high-density lipoprotein-receptor-independent bacterial uptake that is enhanced by CLA-1/CLA-2 overexpression. Bacterial lipopolysaccharides, lipoteichoic acid, and synthetic amphipathic helical peptides (L-37pA and D-37pA) competed with E. coli K12 for CLA-1 and CLA-2 binding. Transmission electron microscopy and confocal microscopy revealed cytosolic accumulation of bacteria in CLA-1/CLA-2-overexpressing HeLa cells. The antibiotic protection assay confirmed that E. coli K12 was able to survive and replicate intracellularly in CLA-1- and CLA-2-overexpressing HeLa, but both L-37pA and D-37pA prevented E. coli K12 invasion. Peritoneal macrophages isolated from SR-BI/BII-knockout mice demonstrated a 30% decrease in bacterial uptake when compared with macrophages from normal mice. Knockout macrophages were also characterized by decreased bacterial cytosolic invasion, ubiquitination, and proteasome mobilization while retaining bacterial lysosomal accumulation. These results indicate that, by facilitating bacterial adhesion and cytosolic invasion, CLA-1 and CLA-2 may play an important role in infection and sepsis.


Journal of Biological Chemistry | 2004

Targeting of scavenger receptor class B type I by synthetic amphipathic α-helical-containing peptides blocks lipopolysaccharide (LPS) uptake and LPS-induced pro-inflammatory cytokine responses in THP-1 monocyte cells

Alexander V. Bocharov; Irina N. Baranova; Tatyana G. Vishnyakova; Alan T. Remaley; Gyorgy Csako; Fairwell Thomas; Amy P. Patterson; Thomas L. Eggerman

Human scavenger receptor class B type I, CLA-1, mediates lipopolysaccharide (LPS) binding and internalization (Vishnyakova, T. G., Bocharov, A. V., Baranova, I. N., Chen, Z., Remaley, A. T., Csako, G., Eggerman, T. L., and Patterson, A. P. (2003) J. Biol. Chem. 278, 22771–22780). Because one of the recognition motifs in SR-B1 ligands is the anionic amphipathic α-helix, we analyzed the effects of model amphipathic α-helical-containing peptides on LPS uptake and LPS-stimulated cytokine production. The L-37pA model peptide, containing two class A amphipathic helices, bound with high affinity (Kd = 0.94 μg/ml) to CLA-1-expressing HeLa cells with a 10-fold increased capacity when compared with mock transfected HeLa cells. Both LPS and L-37pA colocalized with anti-CLA-1 antibody and directly bound CLA-1 as determined by cross-linking. SR-BI/CLA-1 ligands such as HDL, apoA-I, and L-37pA efficiently competed against iodinated L-37pA. Bacterial LPS, lipoteichoic acid, and hsp60 also competed against iodinated L-37pA. Model peptides blocked uptake of iodinated LPS in both mock transfected and CLA-1-overexpressing HeLa cells. Bound and internalized Alexa-L-37pA and BODIPY-LPS colocalized at the cell surface and perinuclear compartment. Both ligands were predominantly transported to the Golgi complex, colocalizing with the Golgi markers bovine serum albumin-ceramide, anti-Golgin97 antibody, and cholera toxin subunit B. A 100-fold excess of L-37pA nearly eliminated BODIPY-LPS binding and internalization. L-37pA and its d-amino acid analogue, D-37pA peptide were similarly effective in blocking LPS, Gram-positive bacterial wall component lipoteichoic acid and bacterial heat shock protein Gro-EL-stimulated cytokine secretion in THP-1 cells. In the same culture media used for the cytokine stimulation study, neither L-37pA nor D-37pA affected the Limulus amebocyte lysate activity of LPS, indicating that LPS uptake and cytokine stimulation were blocked independently of LPS neutralization. These results demonstrate that amphipathic helices of exchangeable apolipoproteins may represent a general host defense mechanism against inflammation.


Journal of Immunology | 2012

Class B Scavenger Receptor Types I and II and CD36 Mediate Bacterial Recognition and Proinflammatory Signaling Induced by Escherichia coli, Lipopolysaccharide, and Cytosolic Chaperonin 60

Irina N. Baranova; Tatyana G. Vishnyakova; Alexander V. Bocharov; Asada Leelahavanichkul; Roger Kurlander; Zhigang Chen; Ana C. P. Souza; Peter S.T. Yuen; Robert A. Star; Gyorgy Csako; Amy P. Patterson; Thomas L. Eggerman

Class B scavenger receptors (SR-B) are lipoprotein receptors that also mediate pathogen recognition, phagocytosis, and clearance as well as pathogen-induced signaling. In this study we report that three members of the SR-B family, namely, CLA-1, CLA-2, and CD36, mediate recognition of bacteria not only through interaction with cell wall LPS but also with cytosolic chaperonin 60. HeLa cells stably transfected with any of these SR-Bs demonstrated markedly (3- to 5-fold) increased binding and endocytosis of Escherichia coli, LPS, and chaperonin 60 (GroEL) as revealed by both FACS analysis and confocal microscopy imaging. Increased pathogen (E. coli, LPS, and GroEL) binding to SR-Bs was also associated with the dose-dependent stimulation of cytokine secretion in the order of CD36 > CLA-2 > CLA-1 in HEK293 cells. Pathogen-induced IL-6-secretion was reduced in macrophages from CD36- and SR-BI/II–null mice by 40–50 and 30–40%, respectively. Intravenous GroEL administration increased plasma IL-6 and CXCL1 levels in mice. The cytokine responses were 40–60% lower in CD36−/− relative to wild-type mice, whereas increased cytokine responses were found in SR-BI/II−/− mice. While investigating the discrepancy of in vitro versus in vivo data in SR-BI/II deficiency, SR-BI/II−/− mice were found to respond to GroEL administration without increases in either plasma corticosterone or aldosterone as normally seen in wild-type mice. SR-BI/II−/− mice with mineralocorticoid replacement demonstrated an ∼40–50% reduction in CXCL1 and IL-6 responses. These results demonstrate that, by recognizing and mediating inflammatory signaling of both bacterial cell wall LPS and cytosolic GroEL, all three SR-B family members play important roles in innate immunity and host defense.


Journal of Lipid Research | 2013

Scavenger receptor-BI is a receptor for lipoprotein(a)

Xiao Ping Yang; Marcelo Amar; Boris Vaisman; Alexander V. Bocharov; Tatyana G. Vishnyakova; Lita Freeman; Roger Kurlander; Amy P. Patterson; Lewis C. Becker; Alan T. Remaley

Scavenger receptor class B type I (SR-BI) is a multi-ligand receptor that binds a variety of lipoproteins, including high density lipoprotein (HDL) and low density lipoprotein (LDL), but lipoprotein(a) [Lp(a)] has not been investigated as a possible ligand. Stable cell lines (HEK293 and HeLa) expressing human SR-BI were incubated with protein- or lipid-labeled Lp(a) to investigate SR-BI-dependent Lp(a) cell association. SR-BI expression enhanced the association of both 125I- and Alexa Fluor-labeled protein from Lp(a). By confocal microscopy, SR-BI was also found to promote the internalization of fluorescent lipids (BODIPY-cholesteryl ester (CE)- and DiI-labeled) from Lp(a), and by immunocytochemistry the cellular internalization of apolipoprotein(a) and apolipoprotein B. When dual-labeled (3H-cholesteryl ether,125I-protein) Lp(a) was added to cells expressing SR-BI, there was a greater relative increase in lipid uptake over protein, indicating that SR-BI mediates selective lipid uptake from Lp(a). Compared with C57BL/6 control mice, transgenic mice overexpressing human SR-BI in liver were found to have increased plasma clearance of 3H-CE-Lp(a), whereas mouse scavenger receptor class B type I knockout (Sr-b1-KO) mice had decreased plasma clearance (fractional catabolic rate: 0.63 ± 0.08/day, 1.64 ± 0.62/day, and 4.64 ± 0.40/day for Sr-b1-KO, C57BL/6, and human scavenger receptor class B type I transgenic mice, respectively). We conclude that Lp(a) is a novel ligand for SR-BI and that SR-BI mediates selective uptake of Lp(a)-associated lipids.


Journal of Immunology | 2012

Class B Scavenger Receptor Types I and II and CD36 Targeting Improves Sepsis Survival and Acute Outcomes in Mice

Asada Leelahavanichkul; Alexander V. Bocharov; Roger Kurlander; Irina N. Baranova; Tatyana G. Vishnyakova; Ana C. P. Souza; Xuzhen Hu; Kent Doi; Boris Vaisman; Marcelo Amar; Denis Sviridov; Zhigang Chen; Alan T. Remaley; Gyorgy Csako; Amy P. Patterson; Peter S.T. Yuen; Robert A. Star; Thomas L. Eggerman

Class B scavenger receptors (SR-Bs), such as SR-BI/II or CD36, bind lipoproteins but also mediate bacterial recognition and phagocytosis. In evaluating whether blocking receptors can prevent intracellular bacterial proliferation, phagocyte cytotoxicity, and proinflammatory signaling in bacterial infection/sepsis, we found that SR-BI/II– or CD36-deficient phagocytes are characterized by a reduced intracellular bacterial survival and a lower cytokine response and were protected from bacterial cytotoxicity in the presence of antibiotics. Mice deficient in either SR-BI/II or CD36 are protected from antibiotic-treated cecal ligation and puncture (CLP)-induced sepsis, with greatly increased peritoneal granulocytic phagocyte survival (8-fold), a drastic diminution in peritoneal bacteria counts, and a 50–70% reduction in systemic inflammation (serum levels of IL-6, TNF-α, and IL-10) and organ damage relative to CLP in wild-type mice. The survival rate of CD36-deficient mice after CLP was 58% compared with 17% in control mice. When compensated for mineralocorticoid and glucocorticoid deficiency, SR-BI/II–deficient mice had nearly a 50% survival rate versus 5% in mineralo-/glucocorticoid-treated controls. Targeting SR-B receptors with L-37pA, a peptide that functions as an antagonist of SR-BI/II and CD36 receptors, also increased peritoneal granulocyte counts, as well as reduced peritoneal bacteria and bacterium-induced cytokine secretion. In the CLP mouse sepsis model, L-37pA improved survival from 6 to 27%, reduced multiple organ damage, and improved kidney function. These results demonstrate that the reduction of both SR-BI/II– and CD36-dependent bacterial invasion and inflammatory response in the presence of antibiotic treatment results in granulocyte survival and local bacterial containment, as well as reduces systemic inflammation and organ damage and improves animal survival during severe infections.


Molecular Therapy | 2014

T-Cell Immunotherapy: Looking Forward

Jacqueline Corrigan-Curay; Hans Peter Kiem; David Baltimore; Marina O'Reilly; Renier J. Brentjens; Laurence J.N. Cooper; Stephen J. Forman; Stephen Gottschalk; Philip D. Greenberg; Richard P. Junghans; Helen E. Heslop; Michael C. Jensen; Crystal L. Mackall; Carl H. June; Oliver W. Press; Daniel J. Powell; Antoni Ribas; Steven A. Rosenberg; Michel Sadelain; Brian G. Till; Amy P. Patterson; Robert Jambou; Eugene Rosenthal; Linda Gargiulo; Maureen Montgomery; Donald B. Kohn

T rapidly expanding field of T-cell immunotherapy has experienced clinical successes along with some serious toxicities. “T Cell Immunotherapy: Optimizing Trial Design,” a workshop sponsored by the National Institutes of Health’s (NIH’s) Office of Biotechnology Activities (OBA), brought together researchers to discuss the scientific advances and share new data on key trial design issues, including the selection of new targets, optimizing the T-cell population, preconditioning regimens, strategies to promote persistence of cells, and analysis and management of acute reactions to T-cell infusions with the goal of identifying best practices and a research agenda that will facilitate further development and maximize the safety of this promising approach.The rapidly expanding field of T-cell immunotherapy has experienced clinical successes along with some serious toxicities. “T Cell Immunotherapy: Optimizing Trial Design,” a workshop sponsored by the National Institutes of Health’s (NIH’s) Office of Biotechnology Activities (OBA), brought together researchers to discuss the scientific advances and share new data on key trial design issues, including the selection of new targets, optimizing the T-cell population, preconditioning regimens, strategies to promote persistence of cells, and analysis and management of acute reactions to T-cell infusions with the goal of identifying best practices and a research agenda that will facilitate further development and maximize the safety of this promising approach.


Science | 2013

A Framework for Decisions About Research with HPAI H5N1 Viruses

Amy P. Patterson; Lawrence A. Tabak; Anthony S. Fauci; Francis S. Collins; Sally Howard

The U.S. Department of Health and Human Services unveils a Framework for funding decisions about highly pathogenic avian influenza H5N1 research. Since it appeared in Hong Kong in 1997, the highly pathogenic avian influenza (HPAI) H5N1 virus has presented a persistent threat to public health and agriculture. Worldwide, hundreds of millions of birds have died as a result of infections or culling to prevent further spread of outbreaks among domestic flocks (1). HPAI H5N1 has caused severe respiratory illness and death in a relatively small number of humans—primarily those who have worked in direct contact with infected poultry (2). Of the ∼600 laboratory-confirmed human cases from 2003 to the present, nearly 60% were fatal. At present, the virus does not appear well-adapted for sustained transmission among mammals by respiratory droplets. However, if the viruses occurring in nature were to become readily transmissible among mammals, they could pose the risk of a pandemic.

Collaboration


Dive into the Amy P. Patterson's collaboration.

Top Co-Authors

Avatar

Thomas L. Eggerman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan T. Remaley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhigang Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gyorgy Csako

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alexander V. Bocharov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Irina N. Baranova

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter S.T. Yuen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ana C. P. Souza

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Roger Kurlander

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge