Network


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

Hotspot


Dive into the research topics where Peter Christmas is active.

Publication


Featured researches published by Peter Christmas.


Journal of Clinical Investigation | 2003

The organization and consequences of eicosanoid signaling

Roy J. Soberman; Peter Christmas

Organization of leukotriene and prostaglandin synthesis As described in the introduction to this Perspective series (1), signaling by arachidonic acid represents a paradigm for the use of oxygen in the transmission of information. At the same time, arachidonic acid signaling can also contribute to the propagation of cellular damage. This duality is typified by a signaling cascade that (a) prevents the activation of 5-lipoxygenase (5-LO) in resting cells and (b) results in the formation and release of leukotrienes (LTs), which requires the sequential activation and interaction of at least eight different proteins. In fact, all lipoxygenases require membrane translocation to exert activity. In the case of the formation of COX products, particularly prostaglandin E2 (PGE2) and PGD2, humans have evolved two sets of biosynthetic enzymes that differ not only in their cell- and tissue-specific localization, but also in their subcellular localization and requirement for reduced glutathione, a cellular defense against oxidative damage. This review will focus on three aspects of arachidonic acid biology. First, the compartmentalization and organization of eicosanoid synthesis, specifically LTs and PGs, will be discussed. This will illustrate the elaborate mechanisms that keep unwanted lipoxygenation at arm’s length and also show that the enzymes such as glutathione-Stransferases, epoxide hydrolases, and carrier proteins that are commonly thought of as biosynthetic also belong to families that are generally considered to play a role in detoxification. Second, the potential cellular oxidative damage that is produced as a by-product of the use of oxygen and lipid substrates is examined. Finally, mechanisms that are used to amplify signaling diversity from a core of LTs and PGs are discussed. The role of leukotrienes C4 and D4 in disease


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

The nuclear membrane organization of leukotriene synthesis

Asim K. Mandal; Phillip B. Jones; Angela M. Bair; Peter Christmas; Douglas K. Miller; Ting-Ting Yamin; Douglas Wisniewski; John G. Menke; Jilly F. Evans; Bradley T. Hyman; Brian J. Bacskai; Mei Chen; D. M. Lee; Boris Nikolic; Roy J. Soberman

Leukotrienes (LTs) are signaling molecules derived from arachidonic acid that initiate and amplify innate and adaptive immunity. In turn, how their synthesis is organized on the nuclear envelope of myeloid cells in response to extracellular signals is not understood. We define the supramolecular architecture of LT synthesis by identifying the activation-dependent assembly of novel multiprotein complexes on the outer and inner nuclear membranes of mast cells. These complexes are centered on the integral membrane protein 5-Lipoxygenase-Activating Protein, which we identify as a scaffold protein for 5-Lipoxygenase, the initial enzyme of LT synthesis. We also identify these complexes in mouse neutrophils isolated from inflamed joints. Our studies reveal the macromolecular organization of LT synthesis.


Journal of Biological Chemistry | 1999

Expression of the CYP4F3 Gene TISSUE-SPECIFIC SPLICING AND ALTERNATIVE PROMOTERS GENERATE HIGH AND LOW Km FORMS OF LEUKOTRIENE B4ω-HYDROXYLASE

Peter Christmas; Sonia R. Ursino; Justin W. Fox; Roy J. Soberman

Cytochrome P450 4F3 (CYP4F3) catalyzes the inactivation of leukotriene B4 by ω-oxidation in human neutrophils. To understand the regulation of CYP4F3 expression, we analyzed the CYP4F3 gene and cloned a novel isoform (CYP4F3B) that is expressed in fetal and adult liver, but not in neutrophils. The CYP4F3 gene contains 14 exons and 13 introns. The cDNAs for CYP4F3A (the neutrophil isoform) and CYP4F3B have identical coding regions, except that they contain exons 4 and 3, respectively. Both exons code for amino acids 66–114 but share only 27% identity. When expressed in COS-7 cells, the K m of CYP4F3B was determined to be 26-fold higher than the K m of CYP4F3A using leukotriene B4 as a substrate. 5′-Rapid amplification of cDNA end studies reveal that the CYP4F3A and CYP4F3B transcripts have 5′-termini derived from different parts of the gene and are initiated from distinct transcription start sites located 519 and 71 base pairs (bp), respectively, from the ATG initiation codon. A consensus TATA box is located 27 bp upstream of the CYP4F3B transcription start site, and a TATA box-like sequence is located 23 bp upstream of the CYP4F3A transcription start site. The data indicate that the tissue-specific expression of functionally distinct CYP4F3 isoforms is regulated by alternative promoter usage and mutually exclusive exon splicing.


Journal of Biological Chemistry | 2002

Membrane localization and topology of leukotriene C4 synthase.

Peter Christmas; Brittany M. Weber; Mary McKee; Dennis Brown; Roy J. Soberman

Leukotriene C4(LTC4) synthase conjugates LTA4 with GSH to form LTC4. Determining the site of LTC4synthesis and the topology of LTC4 synthase may uncover unappreciated intracellular roles for LTC4, as well as how LTC4 is transferred to its export carrier, the multidrug resistance protein-1. We have determined the membrane localization of LTC4 synthase by immunoelectron microscopy. In contrast to the closely related five-lipoxygenase-activating protein, LTC4 synthase is distributed in the outer nuclear membrane and peripheral endoplasmic reticulum but is excluded from the inner nuclear membrane. We have combined immunofluorescence with differential membrane permeabilization to determine the topology of LTC4 synthase. The active site of LTC4 synthase is localized in the lumen of the nuclear envelope and endoplasmic reticulum. These results indicate that the synthesis of LTB4 and LTC4 occurs in different subcellular locations and suggests that LTC4 must be returned to the cytoplasmic side of the membrane for export by multidrug resistance protein-1. The differential localization of two very similar integral membrane proteins suggests that mechanisms other than size-dependent exclusion regulate their passage to the inner nuclear membrane.


PLOS ONE | 2010

20-HETE Mediates Ozone-Induced, Neutrophil-Independent Airway Hyper-Responsiveness in Mice

Philip R. Cooper; A. Clementina Mesaros; Jie Zhang; Peter Christmas; Christopher Stark; Karim Douaidy; Michael Mittelman; Roy J. Soberman; Ian A. Blair; Reynold A. Panettieri

Background Ozone, a pollutant known to induce airway hyper-responsiveness (AHR), increases morbidity and mortality in patients with obstructive airway diseases and asthma. We postulate oxidized lipids mediate in vivo ozone-induced AHR in murine airways. Methodology/Principal Findings Male BALB/c mice were exposed to ozone (3 or 6 ppm) or filtered air (controls) for 2 h. Precision cut lung slices (PCLS; 250 µm thickness) containing an intrapulmonary airway (∼0.01 mm2 lumen area) were prepared immediately after exposure or 16 h later. After 24 h, airways were contracted to carbachol (CCh). Log EC50 and Emax values were then calculated by measuring the airway lumen area with respect to baseline. In parallel studies, dexamethasone (2.5 mg/kg), or 1-aminobenzotriazol (ABT) (50 mg/kg) were given intraperitoneal injection to naïve mice 18 h prior to ozone exposure. Indomethacin (10 mg/kg) was administered 2 h prior. Cell counts, cytokine levels and liquid chromatography-mass spectrometry (LC-MS) for lipid analysis were assessed in bronchoalveolar lavage (BAL) fluid from ozone exposed and control mice. Ozone acutely induced AHR to CCh. Dexamethasone or indomethacin had little effect on the ozone-induced AHR; while, ABT, a cytochrome P450 inhibitor, markedly attenuated airway sensitivity. BAL fluid from ozone exposed animals, which did not contain an increase in neutrophils or interleukin (IL)-6 levels, increased airway sensitivity following in vitro incubation with a naïve PCLS. In parallel, significant increases in oxidized lipids were also identified using LC-MS with increases of 20-HETE that were decreased following ABT treatment. Conclusions/Significance These data show that ozone acutely induces AHR to CCh independent of inflammation and is insensitive to steroid treatment or cyclooxygenase (COX) inhibition. BAL fluid from ozone exposed mice mimicked the effects of in vivo ozone exposure that were associated with marked increases in oxidized lipids. 20-HETE plays a pivotal role in mediating acute ozone-induced AHR.


Journal of Biological Chemistry | 2006

Cytochrome P-450 4F18 Is the Leukotriene B4 ω-1/ω-2 Hydroxylase in Mouse Polymorphonuclear Leukocytes IDENTIFICATION AS THE FUNCTIONAL ORTHOLOGUE OF HUMAN POLYMORPHONUCLEAR LEUKOCYTE CYP4F3A IN THE DOWN-REGULATION OF RESPONSES TO LTB4

Peter Christmas; Karine Tolentino; Valeria Primo; Karin A. Zemski Berry; Robert C. Murphy; Mei Chen; D. M. Lee; Roy J. Soberman

Leukotriene B4 (LTB4) is a potent chemoattractant for polymorphonuclear leukocytes (PMN) and other cells. Human PMN inactivate LTB4 by ω-oxidation catalyzed by cytochrome P-450 (CYP) 4F3A. The contribution of the enzymatic inactivation of LTB4 by CYP4Fs to down-regulating functional responses of cells to LTB4 is unknown. To elucidate the role of CYP4F-mediated inactivation of LTB4 in terminating the responses of PMN to LTB4 and to identify a target for future genetic studies in mice, we have identified the enzyme that catalyzes the ω-1 and ω-2 oxidation of LTB4 in mouse myeloid cells as CYP4F18. As determined by mass spectrometry, this enzyme catalyzes the conversion of LTB4 to 19-OH LTB4 and to a lesser extent 18-OH LTB4. Inhibition of CYP4F18 resulted in a marked increase in calcium flux and a 220% increase in the chemotactic response of mouse PMN to LTB4. CYP4F18 expression was induced in bone marrow-derived dendritic cells by bacterial lipopolysaccharide, a ligand for TLR4, and by poly(I·C), a ligand for TLR3. However, when bone marrow-derived myeloid dendritic cells trafficked to popliteal lymph nodes from paw pads, the expression of CYP4F18 was down-regulated. The results identify CYP4F18 as a critical protein in the regulation of LTB4 metabolism and functional responses in mouse PMN and identify it as the functional orthologue of human PMN CYP4F3A.


Journal of Biological Chemistry | 1999

DIFFERENTIAL LOCALIZATION OF 5- AND 15-LIPOXYGENASES TO THE NUCLEAR ENVELOPE IN RAW MACROPHAGES

Peter Christmas; Justin W. Fox; Sonia R. Ursino; Roy J. Soberman

Leukotriene formation is initiated in myeloid cells by an increase in intracellular calcium and translocation of 5-lipoxygenase from the cytoplasm to the nuclear envelope where it can utilize arachidonic acid. Monocyte- macrophages and eosinophils also express 15-lipoxygenase, which converts arachidonic acid to 15(S)-hydroxyeicosatetraenoic acid. Enhanced green fluorescent 5-lipoxygenase (5-LO) and 15-lipoxygenase (15-LO) fusion proteins were expressed in the cytoplasm of RAW 264.7 macrophages. Only 5-lipoxygenase translocated to the nuclear envelope after cell stimulation, suggesting that differential subcellular compartmentalization can regulate the generation of leukotrienesversus 15(S)-hydroxyeicosatetraenoic acid in cells that possess both lipoxygenases. A series of truncation mutants of 5-LO were created to identify putative targeting domains; none of these mutants localized to the nuclear envelope. The lack of targeting of 15-LO was then exploited to search for specific targeting motifs in 5-LO, by creating 5-LO/15-LO chimeric molecules. The only chimera that could sustain nuclear envelope translocation was one which involved replacement of the N-terminal 237 amino acids with the corresponding segment of 15-LO. Significantly, no discrete targeting domain could be identified in 5-LO, suggesting that sequences throughout the molecule are required for nuclear envelope localization.


PLOS ONE | 2012

CD200R1 Supports HSV-1 Viral Replication and Licenses Pro-Inflammatory Signaling Functions of TLR2

Roy J. Soberman; Christopher R. MacKay; Christine A. Vaine; Glennice Bowen Ryan; Anna M. Cerny; Mikayla R. Thompson; Boris Nikolic; Valeria Primo; Peter Christmas; Paul Sheiffele; Lisa Aronov; David M. Knipe; Evelyn A. Kurt-Jones

The CD200R1:CD200 axis is traditionally considered to limit tissue inflammation by down-regulating pro-inflammatory signaling in myeloid cells bearing the receptor. We generated CD200R1−/− mice and employed them to explore both the role of CD200R1 in regulating macrophage signaling via TLR2 as well as the host response to an in vivo, TLR2-dependent model, herpes simplex virus 1 (HSV-1) infection. CD200R1−/− peritoneal macrophages demonstrated a 70–75% decrease in the generation of IL-6 and CCL5 (Rantes) in response to the TLR2 agonist Pam2CSK4 and to HSV-1. CD200R1−/− macrophages could neither up-regulate the expression of TLR2, nor assemble a functional inflammasome in response to HSV-1. CD200R1−/− mice were protected from HSV-1 infection and exhibited dysfunctional TLR2 signaling. Finally, both CD200R1−/− mice and CD200R1−/− fibroblasts and macrophages showed a markedly reduced ability to support HSV-1 replication. In summary, our data demonstrate an unanticipated and novel requirement for CD200R1 in “licensing” pro-inflammatory functions of TLR2 and in limiting viral replication that are supported by ex vivo and in vivo evidence.


Clinical & Developmental Immunology | 2015

CYP4F18-Deficient Neutrophils Exhibit Increased Chemotaxis to Complement Component C5a

Rachel Vaivoda; Christine A. Vaine; Cassandra Boerstler; Kristy Galloway; Peter Christmas

CYP4Fs were first identified as enzymes that catalyze hydroxylation of leukotriene B4 (LTB4). CYP4F18 has an unusual expression in neutrophils and was predicted to play a role in regulating LTB4-dependent inflammation. We compared chemotaxis of wild-type and Cyp4f18 knockout neutrophils using an in vitro assay. There was no significant difference in the chemotactic response to LTB4, but the response to complement component C5a increased 1.9–2.25-fold in knockout cells compared to wild-type (P < 0.01). This increase was still observed when neutrophils were treated with inhibitors of eicosanoid synthesis. There were no changes in expression of other CYP4 enzymes in knockout neutrophils that might compensate for loss of CYP4F18 or lead to differences in activity. A mouse model of dextran sodium sulfate colitis was used to investigate the consequences of increased C5a-dependent chemotaxis in vivo, but there was no significant difference in weight loss, disease activity, or colonic tissue myeloperoxidase between wild-type and Cyp4f18 knockout mice. This study demonstrates the limitations of inferring CYP4F function based on an ability to use LTB4 as a substrate, points to expanding roles for CYP4F enzymes in immune regulation, and underscores the in vivo challenges of CYP knockout studies.


Biochimica et Biophysica Acta | 2014

Altered leukotriene B4 metabolism in CYP4F18-deficient mice does not impact inflammation following renal ischemia

Valeria Winslow; Rachel Vaivoda; Aleksandr Vasilyev; David Dombkowski; Karim Douaidy; Christopher Stark; Justin Drake; Evin Guilliams; Dharamainder Choudhary; Frederic I. Preffer; Ivaylo Stoilov; Peter Christmas

Inflammatory responses to infection and injury must be restrained and negatively regulated to minimize damage to host tissue. One proposed mechanism involves enzymatic inactivation of the pro-inflammatory mediator leukotriene B4, but it is difficult to dissect the roles of various metabolic enzymes and pathways. A primary candidate for a regulatory pathway is omega oxidation of leukotriene B4 in neutrophils, presumptively by CYP4F3A in humans and CYP4F18 in mice. This pathway generates ω, ω-1, and ω-2 hydroxylated products of leukotriene B4, depending on species. We created mouse models targeting exons 8 and 9 of the Cyp4f18 allele that allows both conventional and conditional knockouts of Cyp4f18. Neutrophils from wild-type mice convert leukotriene B4 to 19-hydroxy leukotriene B4, and to a lesser extent 18-hydroxy leukotriene B4, whereas these products were not detected in neutrophils from conventional Cyp4f18 knockouts. A mouse model of renal ischemia-reperfusion injury was used to investigate the consequences of loss of CYP4F18 in vivo. There were no significant changes in infiltration of neutrophils and other leukocytes into kidney tissue as determined by flow cytometry and immunohistochemistry, or renal injury as assessed by histological scoring and measurement of blood urea nitrogen. It is concluded that CYP4F18 is necessary for omega oxidation of leukotriene B4 in neutrophils, and is not compensated by other CYP enzymes, but loss of this metabolic pathway is not sufficient to impact inflammation and injury following renal ischemia-reperfusion in mice.

Collaboration


Dive into the Peter Christmas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Asim K. Mandal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Boris Nikolic

Albert Einstein Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge