Serveur d'exploration MERS

Attention, ce site est en cours de développement !
Attention, site généré par des moyens informatiques à partir de corpus bruts.
Les informations ne sont donc pas validées.

Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro

Identifieur interne : 001305 ( Istex/Corpus ); précédent : 001304; suivant : 001306

Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro

Auteurs : Ji-Houn Kang ; Geun-Shik Lee ; Eui-Bae Jeung ; Mhan-Pyo Yang

Source :

RBID : ISTEX:3865D293C133D8D879BCDFF7CB068779E57E9D1C

English descriptors

Abstract

Trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.

Url:
DOI: 10.1017/S0007114507280584

Links to Exploration step

ISTEX:3865D293C133D8D879BCDFF7CB068779E57E9D1C

Le document en format XML

<record>
<TEI wicri:istexFullTextTei="biblStruct">
<teiHeader>
<fileDesc>
<titleStmt>
<title xml:lang="en">Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
<author>
<name sortKey="Kang, Ji Houn" sort="Kang, Ji Houn" uniqKey="Kang J" first="Ji-Houn" last="Kang">Ji-Houn Kang</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Lee, Geun Shik" sort="Lee, Geun Shik" uniqKey="Lee G" first="Geun-Shik" last="Lee">Geun-Shik Lee</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Jeung, Eui Bae" sort="Jeung, Eui Bae" uniqKey="Jeung E" first="Eui-Bae" last="Jeung">Eui-Bae Jeung</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Yang, Mhan Pyo" sort="Yang, Mhan Pyo" uniqKey="Yang M" first="Mhan-Pyo" last="Yang">Mhan-Pyo Yang</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
<affiliation>
<mods:affiliation>E-mail: mpyang@chungbuk.ac.kr</mods:affiliation>
</affiliation>
</author>
</titleStmt>
<publicationStmt>
<idno type="wicri:source">ISTEX</idno>
<idno type="RBID">ISTEX:3865D293C133D8D879BCDFF7CB068779E57E9D1C</idno>
<date when="2007" year="2007">2007</date>
<idno type="doi">10.1017/S0007114507280584</idno>
<idno type="url">https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/fulltext.pdf</idno>
<idno type="wicri:Area/Istex/Corpus">001305</idno>
<idno type="wicri:explorRef" wicri:stream="Istex" wicri:step="Corpus" wicri:corpus="ISTEX">001305</idno>
</publicationStmt>
<sourceDesc>
<biblStruct>
<analytic>
<title level="a" type="main" xml:lang="en">Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
<author>
<name sortKey="Kang, Ji Houn" sort="Kang, Ji Houn" uniqKey="Kang J" first="Ji-Houn" last="Kang">Ji-Houn Kang</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Lee, Geun Shik" sort="Lee, Geun Shik" uniqKey="Lee G" first="Geun-Shik" last="Lee">Geun-Shik Lee</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Jeung, Eui Bae" sort="Jeung, Eui Bae" uniqKey="Jeung E" first="Eui-Bae" last="Jeung">Eui-Bae Jeung</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
</author>
<author>
<name sortKey="Yang, Mhan Pyo" sort="Yang, Mhan Pyo" uniqKey="Yang M" first="Mhan-Pyo" last="Yang">Mhan-Pyo Yang</name>
<affiliation>
<mods:affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</mods:affiliation>
</affiliation>
<affiliation>
<mods:affiliation>E-mail: mpyang@chungbuk.ac.kr</mods:affiliation>
</affiliation>
</author>
</analytic>
<monogr></monogr>
<series>
<title level="j">British Journal of Nutrition</title>
<title level="j" type="abbrev">Br J Nutr</title>
<idno type="ISSN">0007-1145</idno>
<idno type="eISSN">1475-2662</idno>
<imprint>
<publisher>Cambridge University Press</publisher>
<pubPlace>Cambridge, UK</pubPlace>
<date type="published" when="2007">2007</date>
<biblScope unit="volume">97</biblScope>
<biblScope unit="issue">1</biblScope>
<biblScope unit="page" from="117">117</biblScope>
<biblScope unit="page" to="125">125</biblScope>
</imprint>
<idno type="ISSN">0007-1145</idno>
</series>
</biblStruct>
</sourceDesc>
<seriesStmt>
<idno type="ISSN">0007-1145</idno>
</seriesStmt>
</fileDesc>
<profileDesc>
<textClass>
<keywords scheme="KwdEn" xml:lang="en">
<term>Conjugated linoleic acid</term>
<term>Neutrophils</term>
<term>Peroxisome proliferator-activated receptor γ</term>
<term>Phagocytosis</term>
<term>Tumour necrosis factor-α</term>
</keywords>
</textClass>
<langUsage>
<language ident="en">en</language>
</langUsage>
</profileDesc>
</teiHeader>
<front>
<div type="abstract" xml:lang="en">Trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.</div>
</front>
</TEI>
<istex>
<corpusName>cambridge</corpusName>
<author>
<json:item>
<name>Ji-Houn Kang</name>
<affiliations>
<json:string>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</json:string>
</affiliations>
</json:item>
<json:item>
<name>Geun-Shik Lee</name>
<affiliations>
<json:string>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</json:string>
</affiliations>
</json:item>
<json:item>
<name>Eui-Bae Jeung</name>
<affiliations>
<json:string>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</json:string>
</affiliations>
</json:item>
<json:item>
<name>Mhan-Pyo Yang</name>
<affiliations>
<json:string>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</json:string>
<json:string>E-mail: mpyang@chungbuk.ac.kr</json:string>
</affiliations>
</json:item>
</author>
<subject>
<json:item>
<lang>
<json:string>eng</json:string>
</lang>
<value>Conjugated linoleic acid</value>
</json:item>
<json:item>
<lang>
<json:string>eng</json:string>
</lang>
<value>Peroxisome proliferator-activated receptor γ</value>
</json:item>
<json:item>
<lang>
<json:string>eng</json:string>
</lang>
<value>Tumour necrosis factor-α</value>
</json:item>
<json:item>
<lang>
<json:string>eng</json:string>
</lang>
<value>Phagocytosis</value>
</json:item>
<json:item>
<lang>
<json:string>eng</json:string>
</lang>
<value>Neutrophils</value>
</json:item>
</subject>
<articleId>
<json:string>01992</json:string>
</articleId>
<arkIstex>ark:/67375/6GQ-7SFQ1NP1-J</arkIstex>
<language>
<json:string>eng</json:string>
</language>
<originalGenre>
<json:string>research-article</json:string>
</originalGenre>
<abstract>Trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.</abstract>
<qualityIndicators>
<score>9.532</score>
<pdfWordCount>5949</pdfWordCount>
<pdfCharCount>36753</pdfCharCount>
<pdfVersion>1.3</pdfVersion>
<pdfPageCount>9</pdfPageCount>
<pdfPageSize>612 x 792 pts (letter)</pdfPageSize>
<refBibsNative>true</refBibsNative>
<abstractWordCount>211</abstractWordCount>
<abstractCharCount>1531</abstractCharCount>
<keywordCount>5</keywordCount>
</qualityIndicators>
<title>Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
<pii>
<json:string>S0007114507280584</json:string>
</pii>
<genre>
<json:string>research-article</json:string>
</genre>
<host>
<title>British Journal of Nutrition</title>
<language>
<json:string>unknown</json:string>
</language>
<issn>
<json:string>0007-1145</json:string>
</issn>
<eissn>
<json:string>1475-2662</json:string>
</eissn>
<publisherId>
<json:string>BJN</json:string>
</publisherId>
<volume>97</volume>
<issue>1</issue>
<pages>
<first>117</first>
<last>125</last>
<total>9</total>
</pages>
<genre>
<json:string>journal</json:string>
</genre>
</host>
<ark>
<json:string>ark:/67375/6GQ-7SFQ1NP1-J</json:string>
</ark>
<categories>
<wos>
<json:string>1 - science</json:string>
<json:string>2 - nutrition & dietetics</json:string>
</wos>
<scienceMetrix>
<json:string>1 - health sciences</json:string>
<json:string>2 - biomedical research</json:string>
<json:string>3 - nutrition & dietetics</json:string>
</scienceMetrix>
<scopus>
<json:string>1 - Health Sciences</json:string>
<json:string>2 - Nursing</json:string>
<json:string>3 - Nutrition and Dietetics</json:string>
<json:string>1 - Health Sciences</json:string>
<json:string>2 - Medicine</json:string>
<json:string>3 - Medicine (miscellaneous)</json:string>
</scopus>
<inist>
<json:string>1 - sciences appliquees, technologies et medecines</json:string>
<json:string>2 - sciences biologiques et medicales</json:string>
<json:string>3 - sciences biologiques fondamentales et appliquees. psychologie</json:string>
</inist>
</categories>
<publicationDate>2007</publicationDate>
<copyrightDate>2007</copyrightDate>
<doi>
<json:string>10.1017/S0007114507280584</json:string>
</doi>
<id>3865D293C133D8D879BCDFF7CB068779E57E9D1C</id>
<score>1</score>
<fulltext>
<json:item>
<extension>pdf</extension>
<original>true</original>
<mimetype>application/pdf</mimetype>
<uri>https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/fulltext.pdf</uri>
</json:item>
<json:item>
<extension>zip</extension>
<original>false</original>
<mimetype>application/zip</mimetype>
<uri>https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/bundle.zip</uri>
</json:item>
<istex:fulltextTEI uri="https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/fulltext.tei">
<teiHeader>
<fileDesc>
<titleStmt>
<title level="a" type="main" xml:lang="en">Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
</titleStmt>
<publicationStmt>
<authority>ISTEX</authority>
<publisher scheme="https://scientific-publisher.data.istex.fr">Cambridge University Press</publisher>
<pubPlace>Cambridge, UK</pubPlace>
<availability>
<licence>
<p>Copyright © The Authors 2007</p>
</licence>
<p scheme="https://loaded-corpus.data.istex.fr/ark:/67375/XBH-G3RCRD03-V">cambridge</p>
</availability>
<date>2007</date>
</publicationStmt>
<notesStmt>
<note type="research-article" scheme="https://content-type.data.istex.fr/ark:/67375/XTP-1JC4F85T-7">research-article</note>
<note type="journal" scheme="https://publication-type.data.istex.fr/ark:/67375/JMC-0GLKJH51-B">journal</note>
<note>Abbreviations: BADGE, bisphenol A diglycidyl ether; CLA, conjugated linoleic acid; FITC, fluorescein isothiocyanate; PAb, polyclonal antibody; PBMC, peripheral blood mononuclear cells; PMN, polymorphonuclear cells; rh, recombinant human; rp, recombinant porcine; t10c12, trans-10, cis-12</note>
</notesStmt>
<sourceDesc>
<biblStruct type="inbook">
<analytic>
<title level="a" type="main" xml:lang="en">Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
<author xml:id="author-0000">
<persName>
<forename type="first">Ji-Houn</forename>
<surname>Kang</surname>
</persName>
<affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
</author>
<author xml:id="author-0001">
<persName>
<forename type="first">Geun-Shik</forename>
<surname>Lee</surname>
</persName>
<affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
</author>
<author xml:id="author-0002">
<persName>
<forename type="first">Eui-Bae</forename>
<surname>Jeung</surname>
</persName>
<affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
</author>
<author xml:id="author-0003" corresp="yes">
<persName>
<forename type="first">Mhan-Pyo</forename>
<surname>Yang</surname>
</persName>
<email>mpyang@chungbuk.ac.kr</email>
<affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
</author>
<idno type="istex">3865D293C133D8D879BCDFF7CB068779E57E9D1C</idno>
<idno type="ark">ark:/67375/6GQ-7SFQ1NP1-J</idno>
<idno type="DOI">10.1017/S0007114507280584</idno>
<idno type="PII">S0007114507280584</idno>
<idno type="article-id">01992</idno>
</analytic>
<monogr>
<title level="j">British Journal of Nutrition</title>
<title level="j" type="abbrev">Br J Nutr</title>
<idno type="pISSN">0007-1145</idno>
<idno type="eISSN">1475-2662</idno>
<idno type="publisher-id">BJN</idno>
<imprint>
<publisher>Cambridge University Press</publisher>
<pubPlace>Cambridge, UK</pubPlace>
<date type="published" when="2007"></date>
<biblScope unit="volume">97</biblScope>
<biblScope unit="issue">1</biblScope>
<biblScope unit="page" from="117">117</biblScope>
<biblScope unit="page" to="125">125</biblScope>
</imprint>
</monogr>
</biblStruct>
</sourceDesc>
</fileDesc>
<profileDesc>
<creation>
<date>2007</date>
</creation>
<langUsage>
<language ident="en">en</language>
</langUsage>
<abstract xml:lang="en" style="normal">
<p>Trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.</p>
</abstract>
<textClass xml:lang="en">
<keywords scheme="keyword">
<list>
<head></head>
<item>
<term>Conjugated linoleic acid</term>
</item>
<item>
<term>Peroxisome proliferator-activated receptor γ</term>
</item>
<item>
<term>Tumour necrosis factor-α</term>
</item>
<item>
<term>Phagocytosis</term>
</item>
<item>
<term>Neutrophils</term>
</item>
</list>
</keywords>
</textClass>
</profileDesc>
<revisionDesc>
<change when="2007">Published</change>
</revisionDesc>
</teiHeader>
</istex:fulltextTEI>
<json:item>
<extension>txt</extension>
<original>false</original>
<mimetype>text/plain</mimetype>
<uri>https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/fulltext.txt</uri>
</json:item>
</fulltext>
<metadata>
<istex:metadataXml wicri:clean="corpus cambridge not found" wicri:toSee="no header">
<istex:xmlDeclaration>version="1.0" encoding="UTF-8"</istex:xmlDeclaration>
<istex:docType PUBLIC="-//NLM//DTD Journal Publishing DTD v2.2 20060430//EN" URI="journalpublishing.dtd" name="istex:docType"></istex:docType>
<istex:document>
<article article-type="research-article" dtd-version="2.2" xml:lang="EN">
<front>
<journal-meta>
<journal-id journal-id-type="publisher-id">BJN</journal-id>
<journal-title>British Journal of Nutrition</journal-title>
<abbrev-journal-title>Br J Nutr</abbrev-journal-title>
<issn pub-type="ppub">0007-1145</issn>
<issn pub-type="epub">1475-2662</issn>
<publisher>
<publisher-name>Cambridge University Press</publisher-name>
<publisher-loc>Cambridge, UK</publisher-loc>
</publisher>
</journal-meta>
<article-meta>
<article-id pub-id-type="doi">10.1017/S0007114507280584</article-id>
<article-id pub-id-type="pii">S0007114507280584</article-id>
<article-id pub-id-type="publisher-id">01992</article-id>
<title-group>
<article-title>
<italic>Trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells
<italic>in vitro</italic>
</article-title>
<alt-title alt-title-type="left-running">Conjugated linoleic acid and phagocytosis</alt-title>
<alt-title alt-title-type="right-running">J.-H. Kang
<italic>et al.</italic>
</alt-title>
</title-group>
<contrib-group>
<contrib contrib-type="author">
<name name-style="western">
<surname>Kang</surname>
<given-names>Ji-Houn</given-names>
</name>
<xref ref-type="aff" rid="aff001">
<sup>1</sup>
</xref>
</contrib>
<contrib contrib-type="author">
<name name-style="western">
<surname>Lee</surname>
<given-names>Geun-Shik</given-names>
</name>
<xref ref-type="aff" rid="aff002">
<sup>2</sup>
</xref>
</contrib>
<contrib contrib-type="author">
<name name-style="western">
<surname>Jeung</surname>
<given-names>Eui-Bae</given-names>
</name>
<xref ref-type="aff" rid="aff002">
<sup>2</sup>
</xref>
</contrib>
<contrib contrib-type="author" corresp="yes">
<name name-style="western">
<surname>Yang</surname>
<given-names>Mhan-Pyo</given-names>
</name>
<xref ref-type="aff" rid="aff001">
<sup>1</sup>
</xref>
<xref ref-type="corresp" rid="cor001">*</xref>
</contrib>
</contrib-group>
<aff id="aff001">
<label>
<sup>1</sup>
</label>
<addr-line>Laboratory of Veterinary Internal Medicine</addr-line>
,
<addr-line>Department of Veterinary Medicine</addr-line>
,
<institution>College of Veterinary Medicine and Research Institute of Veterinary Medicine</institution>
,
<addr-line>Chungbuk National University</addr-line>
,
<addr-line>Cheongju</addr-line>
,
<addr-line>Chungbuk 361-763</addr-line>
,
<country>South Korea</country>
</aff>
<aff id="aff002">
<label>
<sup>2</sup>
</label>
<addr-line>Laboratory of Veterinary Biochemistry and Molecular Biology</addr-line>
,
<addr-line>Department of Veterinary Medicine</addr-line>
,
<institution>College of Veterinary Medicine and Research Institute of Veterinary Medicine</institution>
,
<addr-line>Chungbuk National University</addr-line>
,
<addr-line>Cheongju</addr-line>
,
<addr-line>Chungbuk 361-763</addr-line>
,
<country>South Korea</country>
</aff>
<author-notes>
<corresp id="cor001">
<bold>*</bold>
<bold>Corresponding author:</bold>
Dr Mhan-Pyo Yang, fax +82 43 261 3224, email
<email xlink:href="mpyang@chungbuk.ac.kr">mpyang@chungbuk.ac.kr</email>
</corresp>
</author-notes>
<pub-date pub-type="epub-ppub">
<month>01</month>
<year>2007</year>
</pub-date>
<volume>97</volume>
<issue>1</issue>
<fpage>117</fpage>
<lpage>125</lpage>
<history>
<date date-type="received">
<day>11</day>
<month>04</month>
<year>2006</year>
</date>
<date date-type="rev-recd">
<day>23</day>
<month>08</month>
<year>2006</year>
</date>
<date date-type="accepted">
<day>18</day>
<month>09</month>
<year>2006</year>
</date>
</history>
<permissions>
<copyright-statement>Copyright © The Authors 2007</copyright-statement>
<copyright-year>2007</copyright-year>
<copyright-holder>The Authors</copyright-holder>
</permissions>
<abstract abstract-type="normal">
<p>
<italic>Trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.</p>
</abstract>
<kwd-group kwd-group-type="" xml:lang="en">
<kwd>Conjugated linoleic acid</kwd>
<kwd>Peroxisome proliferator-activated receptor γ</kwd>
<kwd>Tumour necrosis factor-α</kwd>
<kwd>Phagocytosis</kwd>
<kwd>Neutrophils</kwd>
</kwd-group>
<counts>
<page-count count="9"></page-count>
</counts>
<custom-meta-wrap>
<custom-meta>
<meta-name>pdf</meta-name>
<meta-value>S0007114507280584a.pdf</meta-value>
</custom-meta>
</custom-meta-wrap>
</article-meta>
<notes>
<p>
<bold>Abbreviations:</bold>
BADGE, bisphenol A diglycidyl ether; CLA, conjugated linoleic acid; FITC, fluorescein isothiocyanate; PAb, polyclonal antibody; PBMC, peripheral blood mononuclear cells; PMN, polymorphonuclear cells; rh, recombinant human; rp, recombinant porcine; t10c12,
<italic>trans</italic>
-10,
<italic>cis</italic>
-12</p>
</notes>
</front>
<body>
<p>PPAR represent a subfamily of nuclear hormone receptors that are activated by a variety of dietary and endogenous fatty acids (Schoonjans
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref42">1996</xref>
; Xu
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref51">1999</xref>
). The PPAR family is currently divided into three subgroups: α, β/δ, and γ (Daynes & Jones,
<xref ref-type="bibr" rid="ref11">2002</xref>
). These subgroups are characterised by distinct patterns of tissue distribution and metabolic function (Braissant
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref5">1996</xref>
; Schoonjans
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref41">1997</xref>
). Some PPARγ agonists, such as 15-deoxy-Δ
<sup>12,14</sup>
-prostaglandin J
<sub>2</sub>
and the insulin-sensitising thiazolidinediones, have been reported to exert anti-inflammatory actions by reducing inflammation-associated molecules (Ricote
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref38">1998</xref>
). The PPARγ agonists, 15-deoxy-Δ
<sup>12,14</sup>
-prostaglandin J
<sub>2</sub>
and troglitazone, inhibit phorbol myristyl acetate-induced production of IL-1β, IL-6, and TNF-α in peripheral blood monocytes (Jiang
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref21">1998</xref>
). However, pioglitazone and rosiglitazone, which are also PPARγ agonists, do not inhibit phorbol ester-induced TNF-α release from human monocytic THP-1 cells (Cunard
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref10">2002</xref>
). Although PPARγ was originally shown to play a key role in adipocyte differentiation and lipid metabolism (Chawla
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref9">1994</xref>
), it potentially may play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages.</p>
<p>In clinical medicine, nutritional therapy may be important for immunoregulation, inflammation, neoplasia and vascular diseases (Calder,
<xref ref-type="bibr" rid="ref8">1998</xref>
). Modulation of the immune system by dietary fatty acids may occur through regulation of arachidonic acid metabolism and eicosanoid production (Calder,
<xref ref-type="bibr" rid="ref7">1996</xref>
; Miles
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref31">2002</xref>
), modification of membrane fluidity (Mexmain
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref30">1984</xref>
) and transcriptional regulation of gene expression by PPAR (Bishop-Bailey & Wray,
<xref ref-type="bibr" rid="ref4">2003</xref>
). Conjugated linoleic acid (CLA), a dietary fatty acid, has received special attention since it can trigger modification of immune cell functions, as well as having numerous other physiological characteristics such as anti-adipogenic (Smedman & Vessby,
<xref ref-type="bibr" rid="ref46">2001</xref>
), anti-diabetogenic (Ryder
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref40">2001</xref>
), anti-carcinogenic (Palombo
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref34">2002</xref>
) and anti-atherosclerotic (Lee
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref26">1994</xref>
) properties. Recently, the putative detrimental influences of CLA have been reported. CLA acts as a cancer promoter in colon carcinogenesis (Rajakangas
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref37">2003</xref>
) and increases inflammatory indicators such as C-reactive protein (Smedman
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref45">2005</xref>
). It causes significant impairment of peripheral insulin sensitivity as well as of blood glucose and serum lipid concentrations (Risérus
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref39">2004</xref>
).</p>
<p>CLA can stimulate or inhibit immune cell function. CLA increases TNF-α and IL-6 secretion and decreases IL-4 secretion by splenocytes (Kelley
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref23">2002</xref>
). Expression of PPARγ is enhanced in CLA-fed pigs (Meadus
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref29">2002</xref>
). CLA is capable of modulating pro-inflammatory signals such as TNF-α and inducible NO synthase in macrophages. The regulation of inducible NO synthase transcription by CLA requires PPARγ, as demonstrated using a dominant negative construct (Yu
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref54">2002</xref>
). CLA treatment ameliorates the symptoms of type α diabetes mellitus in Zucker diabetic fa/fa rats and affects differentiation of adipose tissue (Houseknecht
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref17">1998</xref>
). In contrast, CLA down regulates the expression of PPARγ and its target genes, fatty-acid binding protein, and liver X receptor α in adipocytes (Granlund
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref14">2003</xref>
). These reports indicate that the effects of CLA in modulating immune responses are similar to effects induced by ligands of PPARγ.</p>
<p>In particular,
<italic>trans</italic>
-10,
<italic>cis</italic>
-12 (t10c12)-CLA, a CLA isomer, is known to alter immune function (Pariza
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref35">2001</xref>
). In previous studies, it has been shown that direct treatment with t10c12-CLA has no effect on the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN). However, the phagocytic capacity of porcine PMN is markedly enhanced by the culture supernatant fraction from porcine peripheral blood mononuclear cells (PBMC) treated with t10c12-CLA (Kang
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref22">2004</xref>
). Therefore, it has been hypothesised that t10c12-CLA involves the production of soluble factor(s) including TNF-α, a phagocytosis-promoting factor, from porcine PBMC that enhance the phagocytic capacity of porcine PMN, which may be an important mechanism for the enhancement of the innate immune response.</p>
<p>In the present study, we examined whether t10c12-CLA stimulates PPARγ expression in porcine PBMC and, if so, whether activation of PPARγ in PBMC by t10c12-CLA affects TNF-α production, which enhances the phagocytic capacity of PMN. In addition, the effects of PPARγ antagonism on TNF-α production by PBMC and the phagocytic capacity of PMN were examined.</p>
<sec id="sec1" sec-type="materialsandmethods">
<title>Materials and methods</title>
<sec id="sec1-1">
<title>Pigs</title>
<p>Healthy 5-month-old Landrace crossbred pigs were used as blood donors. Pigs were kept in a temperature-controlled room with a 12 h light–dark cycle and fed a commercial diet (Fildmaster; Purina Korea, Seoul, Korea) and tap water. All experimental procedures and animal use were approved by the ethics committee of the Chungbuk National University (South Korea).</p>
</sec>
<sec id="sec1-2">
<title>Reagents and antibodies</title>
<p>t10c12-CLA (98 % purity) was purchased commercially (Matreya Inc., Pleasant Gap, PA, USA). t10c12-CLA stock solution was prepared by dissolving t10c12-CLA in dimethyl sulfoxide (Moya-Camarena
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref32">1999</xref>
; Brown
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref6">2003</xref>
) to a final concentration of 50 m
<sc>m</sc>
and passed through a 0·45 μm membrane filter (Millipore Co., Bedford, MA, USA) before use. Recombinant porcine (rp) TNF-α, goat anti-rpTNF-α polyclonal antibody (pAb) (R&D Systems Inc., Minneapolis, MN, USA), goat anti-recombinant human (rh) IL-2 pAb (Sigma-Aldrich Co., St Louis, MO, USA), and bisphenol A diglycidyl ether (BADGE) (Fluka Chemie AG, Buchs, Switzerland) were purchased commercially.</p>
</sec>
<sec id="sec1-3">
<title>Isolation of peripheral blood mononuclear cells and polymorphonuclear cells</title>
<p>Heparinised porcine peripheral blood was drawn from the anterior vena cava and diluted with an equal volume of PBS without Ca and Mg and overlayed 1:1 on a Histopaque solution (specific gravity, 1·080; Sigma-Aldrich Co.). After centrifugation at 400 
<italic>
<bold>g</bold>
</italic>
for 45 min at room temperature, the cells in the interface between the plasma in PBS and the Histopaque solution were harvested and treated with 0·83 % NH
<sub>4</sub>
Cl in a tri(hydroxymethyl)-aminomethane-base buffer (pH 7·2) for 5 min. The resulting PBMC were washed three times with PBS. PMN were obtained from the upper layer of sedimented erythrocytes after the removal of the PBMC layer. The erythrocytes were allowed to sediment for 60 min with dextran (molecular weight, 200 000; Wako Ltd, Osaka, Japan). The floating cells were gently collected and pelleted by centrifugation at 400 
<italic>
<bold>g</bold>
</italic>
for 5 min. The residual erythrocytes were lysed by transitory treatment with 0·83 % NH
<sub>4</sub>
Cl. The purity of neutrophils in the final PMN suspension was routinely greater than 95 % as determined by cytospin smear and Wright–Giemsa stain. Both PBMC and PMN were re-suspended in RPMI 1640 medium (Sigma-Aldrich Co.) supplemented with 2 m
<sc>m</sc>
-
<sc>l</sc>
-glutamine, gentamicin (0·02 mg/ml), and 5 % heat-inactivated fetal bovine serum (Invitrogen Co., Grand Island, NY, USA).</p>
</sec>
<sec id="sec1-4">
<title>Treatment of peripheral blood mononuclear cells</title>
<p>t10c12-CLA and/or BADGE (100 μ
<sc>m</sc>
), a PPARγ antagonist, were added to freshly isolated PBMC culture media with a minimal volume ( <  0·1 %) of dimethyl sulfoxide as the solvent and the same amount as vehicle dimethyl sulfoxide was added to control cells. To determine mRNA expression in cells, PBMC at a density of 2 × 10
<sup>6</sup>
cells/ml per well in a twenty-four-well plate (Nunc Co., Naperville, IL, USA) were incubated for 3 h at 37°C under a 5 % CO
<sub>2</sub>
-humidified atmosphere. After incubation, the samples were aspirated and centrifuged at 400 
<italic>
<bold>g</bold>
</italic>
for 30 min at 4°C. The pelleted PBMC were stored at − 70°C until use in further analysis. The PBMC were also incubated for 24 h to obtain the PBMC culture supernatant fraction. The supernatant fractions were centrifuged at 5000 
<italic>
<bold>g</bold>
</italic>
for 30 min at 4°C, filtered with a 0·45 μm pore membrane filter, and stored at − 70°C. At the end of the incubation period, the viability of cells was consistently more than 95 % as determined by trypan blue dye exclusion.</p>
</sec>
<sec id="sec1-5">
<title>Phagocytosis assay</title>
<p>PMN (100 μl), adjusted to 1 × 10
<sup>7</sup>
cells/ml, were added to each well of a twenty-four-well plate. The PMN were incubated with the culture supernatant fraction from PBMC that had been treated with either various concentrations of t10c12-CLA or t10c12-CLA in combination with BADGE (100 μ
<sc>m</sc>
) for 12 h at 37°C under a 5 % CO
<sub>2</sub>
-humidified atmosphere. Fluorescein isothiocyanate (FITC)-latex beads (20 μl; 1 × 10
<sup>9</sup>
beads/ml; bead size 2·0 μm; Polysciences Inc., Warrington, PA, USA) were added to each well for the final 1 h. PMN incubated without FITC-latex beads were used as a negative control. The cultured cells were gently harvested, centrifuged at 400 
<italic>
<bold>g</bold>
</italic>
for 3 min at 4°C and washed three times with PBS containing 3 m
<sc>m</sc>
-EDTA. After washing three times, the supernatant fraction was discarded and replaced with PBS containing 1 % paraformaldehyde to stabilise the cells. Surface-adherent FITC fluorescence on PMN was quenched by the addition of 20 μl of 0·4 % trypan blue solution to each tube before flow cytometry analysis as previously described (Hed
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref16">1987</xref>
). The use of trypan blue solution has an effect in fluorescence quenching on attached particles but not ingested particles (Antal-Szalmás
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref1">2000</xref>
). Phagocytosis was measured by FACS Calibur (Becton Dickinson Immunocytometry Systems, San Jose, CA, USA) flow cytometry using CELLQuest software. The Ar laser was set to emit an excitation wavelength of 488 nm. FITC (green) fluorescence was measured between 515 and 560 nm on 10 000 PMN per sample. PMN were gated by forward and side light scatter characteristics. The PMN were monitored by propidium iodide (Sigma-Aldrich Co.) for live cells gated by flow cytometry. The results are expressed as percentages of absolute phagocytic capacity.</p>
</sec>
<sec id="sec1-6">
<title>Neutralisation test</title>
<p>For the neutralisation test, anti-rpTNF pAb was diluted to various concentrations and added to the t10c12-CLA-stimulated PBMC culture supernatant fraction. Goat anti-rhIL-2 pAb was used as a control isotype IgG. The mixed samples were kept for 30 min at room temperature.</p>
</sec>
<sec id="sec1-7">
<title>Measurement of tumour necrosis factor-α in the peripheral blood mononuclear cells culture supernatant fraction</title>
<p>Porcine PBMC were incubated with t10c12-CLA alone or t10c12-CLA plus BADGE (100 μ
<sc>m</sc>
). The culture supernatant fraction was collected and the amount of TNF-α was determined by a direct sandwich ELISA using the Quantikine
<sup>®</sup>
P porcine TNF-α immunoassay kit (R&D Systems Inc.) according to the manufacturer's protocol. All samples, standards and controls were assayed in triplicate. The optical density was determined using an automated microplate reader (EL × 808; BioTek Instruments Inc., Winooski, VT, USA) at 450 nm. TNF-α was quantified from eight titration points using standard curves generated with purified porcine TNF-α, and the concentrations were expressed as pg/ml. Lower and upper detection limits were 11·7 and 1500 pg/ml, respectively.</p>
</sec>
<sec id="sec1-8">
<title>Ribonucleic acid preparation and reverse transcriptase-polymerase chain reaction</title>
<p>Total RNA was prepared from porcine PBMC according to a protocol for single-step RNA isolation based on acid guanidinium-thiocyanate-phenol-chloroform extraction, using TRIzol reagent (Invitrogen Co., Carlsbad, CA, USA) as previously described (Yang
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref53">2002</xref>
). Total RNA (2 μg) was reverse transcribed into cDNA using the Moloney-murine leukaemia virus RT (Ambion Inc., Austin, TX, USA) and random primers (9-mers). To determine the conditions under which PCR amplification of PPARγ, TNF-α and cytochrome c oxidase subunit (1A) mRNA were in the logarithmic phase, samples (1 μl) were amplified using different numbers of cycles. The 1A gene was PCR-amplified to rule out the possibility of RNA degradation and to control for variations in mRNA concentrations in the RT reaction. PCR products and amplification cycles were linearly related for PPARγ, TNF-α and 1A mRNA. Thirty cycles for PPARγ and TNF-α and twenty-five cycles for 1A were employed for quantification. The cDNA were amplified in 20 μl PCR reactions containing 1 unit
<italic>Taq</italic>
polymerase (Promega Co., Madison, WI, USA) and its buffer, 1·5 m
<sc>m</sc>
-MgCl
<sub>2</sub>
, 2 m
<sc>m</sc>
dNTP and 50 pmol of specific primers. PCR reactions were denatured at 95°C for 1 min, annealed at 50°C for 1 min, and extended at 72°C for 1 min, 30 s. cDNA sequences of mRNA were obtained by RT reaction primers (Macrogen Inc., Seoul, Korea). The oligonucleotides for PPARγ were based on the cDNA sequence (GenBank accession number AJ006756): 5′-CTG GCA AAG CAC TTG TAT G-3′ (sense) and 5′-GGT GTA AAT GAT CTC GTG GA-3′ (anti-sense). The oligonucleotides for TNF-α were based on the cDNA sequence (GenBank accession number X57321): 5′-CAA GGA CTC AGA TCA TCG TC-3′ (sense) and 5′-CTT GGT CTG GTA GGA GAC G-3′ (anti-sense). The primers for the 1A gene (GenBank accession number AF03253) were 5′-CAC CGT AGG AGG TCT AAC G-3′ (sense) and 5′-GTA TCG TCG AGG TAT TCC G-3′ (anti-sense). Of the PCR products, 10 μl were fractionated on a 2 % agarose gel and stained with ethidium bromide. The photograph was scanned and analysed using the molecular analysis program Gel Doc 1000 version 1.5 (Bio-Rad Lab., Hercules, CA, USA).</p>
</sec>
<sec id="sec1-9">
<title>Statistical analysis</title>
<p>All statistical analyses were carried out using the software SigmaStat version 2.03 (SPSS Inc., Chicago, IL, USA) or SAS version 9.1 (SAS Institute Co., Cary, NC, USA). One-way ANOVA was used to investigate differences between control and concentrations of CLA treatment, followed by Dunnett's
<italic>post hoc</italic>
test. Comparisons of two groups at each CLA concentration were made by Student's
<italic>t</italic>
test, separately.
<italic>P</italic>
 < 0·05 was considered statistically significant. Data were expressed as mean values and standard deviations.</p>
</sec>
</sec>
<sec id="sec2" sec-type="results">
<title>Results</title>
<sec id="sec2-1">
<title>Effect of trans-10, cis-12-conjugated linoleic acid on tumour necrosis factor-α expression and production in porcine peripheral blood mononuclear cells</title>
<p>TNF-α mRNA expression in PBMC in response to t10c12-CLA was examined. The isolated PBMC were treated with t10c12-CLA (2·5–20 μ
<sc>m</sc>
) and harvested for total RNA extraction 3 h later. TNF-α mRNA expression in PBMC was slightly increased by t10c12-CLA and peaked at a concentration of 5 μ
<sc>m</sc>
(
<xref ref-type="fig" rid="fig1">Fig. 1 (A)</xref>
). t10c12-CLA induced a significant increase (
<italic>P</italic>
 < 0·001) in TNF-α production by PBMC in a dose-dependent manner (
<xref ref-type="fig" rid="fig1">Fig. 1 (B)</xref>
).
<fig id="fig1" position="float">
<label>Fig. 1</label>
<caption>
<p>TNF-α expression and production in porcine peripheral blood mononuclear cells (PBMC) by
<italic>trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid (t10c12-CLA). (A) RT-PCR analysis of TNF-α mRNA expression in porcine PBMC treated with t10c12-CLA at the indicated concentrations for 3 h (a). Normalisation of TNF-α mRNA expression with 1A (b). Signals were quantified with a molecular analysis program and expressed as a percentage of the maximum values (c). The expected product sizes of TNF-α and 1A mRNA are 273 and 277 bp, respectively. (B) The amount of TNF-α in the porcine PBMC culture supernatant fraction treated with t10c12-CLA at the indicated concentrations for 24 h was determined using an ELISA (see p. 118). One-way ANOVA was used to investigate differences between control and concentrations of CLA treatment, followed by Dunnett's
<italic>post hoc</italic>
test. Data are means (
<italic>n</italic>
6), with standard deviations represented by vertical bars. * Mean value was significantly different from that for the vehicle treatment (0 μ
<sc>m</sc>
-t10c12-CLA) (
<italic>P</italic>
 < 0·05).</p>
</caption>
<graphic alt-version="no" mime-subtype="gif" mimetype="image" position="float" xlink:href="S0007114507280584_fig1"></graphic>
</fig>
</p>
</sec>
<sec id="sec2-2">
<title>Tumour necrosis factor-α increases the phagocytic capacity of porcine polymorphonuclear cells</title>
<p>To examine the phagocytic capacity of PMN exposed to the culture supernatant fraction from PBMC treated with t10c12-CLA, PMN were incubated for 12 h with the culture supernatant fraction from PBMC treated with 2·5–20 μ
<sc>m</sc>
-t10c12-CLA. The phagocytic capacity of PMN was significantly enhanced (
<italic>P</italic>
 < 0·001) in a dose-dependent manner by the culture supernatant fraction (
<xref ref-type="fig" rid="fig2">Fig. 2 (A)</xref>
). Its capacity was also significantly increased (
<italic>P</italic>
 < 0·001) in a dose-dependent manner by the addition of rpTNF-α (
<xref ref-type="fig" rid="fig2">Fig. 2 (B)</xref>
). Anti-rpTNF-α pAb neutralised the enhancement of PMN phagocytic capacity by the PBMC culture supernatant fraction treated with t10c12-CLA (10 μ
<sc>m</sc>
) (
<italic>P</italic>
 < 0·001) in a dose-dependent manner (
<xref ref-type="fig" rid="fig2">Fig. 2 (C)</xref>
). There was no non-specific inhibition by an immunoglobulin IgG isotype of anti-rpTNF-α pAb, since the enhanced phagocytic capacity was not inhibited by the addition of a high concentration (1 μg/ml) of the anti-rhIL-2 pAb used as a control IgG.
<fig id="fig2" position="float">
<label>Fig. 2</label>
<caption>
<p>The phagocytic capacity of porcine polymorphonuclear cells (PMN). (A) The effect of the culture supernatant fraction from porcine peripheral blood mononuclear cells (PBMC) treated with
<italic>trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid (t10c12-CLA) on phagocytic capacity of PMN. Freshly isolated PMN (1 × 10
<sup>6</sup>
cells/ml per well) were incubated for 12 h with the culture supernatant fraction from PBMC (2 × 10
<sup>6</sup>
cells/ml) that had been treated with t10c12-CLA at the indicated concentrations for 24 h. One-way ANOVA was used to investigate differences between control and treatments, followed by Dunnett's
<italic>post hoc</italic>
test. Data are means (
<italic>n</italic>
6), with standard deviations represented by vertical bars. * Mean value was significantly different from that for the vehicle (0 μ
<sc>m</sc>
-t10c12-CLA)-treated PBMC culture supernatant fraction (
<italic>P</italic>
 < 0·05). (B) The effect of recombinant porcine (rp) TNF-α on the phagocytic capacity of PMN. PMN (1 × 10
<sup>6</sup>
cells/ml per well) were treated with rpTNF-α at the indicated concentrations for 12 h. The culture supernatant fraction from PBMC (2 × 10
<sup>6</sup>
cells/ml) treated with t10c12-CLA (10 μ
<sc>m</sc>
) for 24 h was prepared as a positive control. One-way ANOVA was used to investigate differences between control and treatments, followed by Dunnett's
<italic>post hoc</italic>
test. Data are means (
<italic>n</italic>
6), with standard deviations represented by horizontal bars. * Mean value was significantly different from that for the vehicle-treated PBMC culture supernatant fraction (
<italic>P</italic>
 < 0·05). (C) The neutralising effect of anti-rpTNF-α polyclonal antibody (pAb) on the phagocytic capacity of PMN. Anti-rpTNF-α pAb, at the indicated concentrations, was added to the culture supernatant fraction from PBMC (2 × 10
<sup>6</sup>
cells/ml) treated with t10c12-CLA (10 μ
<sc>m</sc>
) for 24 h. Goat anti-recombinant human IL-2 pAb (1 μg/ml) was used as a control isotype IgG. The mixed samples were kept for 30 min. Fluorescein isothiocyanate-latex beads were added to all cultures for the final 1 h. The phagocytic capacity of PMN was measured using flow cytometry (see pp. 118-119). One-way ANOVA was used to investigate differences between control and treatments, followed by Dunnett's
<italic>post hoc</italic>
test. Data are means (
<italic>n</italic>
6), with standard deviations represented by horizontal bars. * Mean value was significantly different from that for the t10c12-CLA-treated PBMC culture supernatant fraction (
<italic>P</italic>
 < 0·05).</p>
</caption>
<graphic alt-version="no" mime-subtype="gif" mimetype="image" position="float" xlink:href="S0007114507280584_fig2"></graphic>
</fig>
</p>
</sec>
<sec id="sec2-3">
<title>Effect of trans-10, cis-12-conjugated linoleic acid on peroxisome proliferator-activated receptor-γ mRNA expression in porcine peripheral blood mononuclear cells</title>
<p>To examine the expression of PPARγ mRNA in PBMC in response to t10c12-CLA, PBMC were incubated with t10c12-CLA and harvested for RNA isolation. Porcine PPARγ mRNA expression in PBMC was increased in a dose-dependent manner by t10c12-CLA treatment and peaked at 5 μ
<sc>m</sc>
-t10c12-CLA, although a detectable PPARγ signal was seen in the vehicle-treated PBMC after 3 h of incubation (
<xref ref-type="fig" rid="fig3">Fig. 3</xref>
).
<fig id="fig3" position="float">
<label>Fig. 3</label>
<caption>
<p>PPARγ mRNA expression in porcine peripheral blood mononuclear cells (PBMC) after
<italic>trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid (t10c12-CLA) treatment. RT-PCR analysis of PPARγ mRNA expression in PBMC treated with t10c12-CLA at the indicated concentrations for 3 h (a). Normalisation of the PPARγ mRNA expression with 1A (b). Signals were quantified with a molecular analysis program and expressed as a percentage of the maximum values (c). The expected product sizes of PPARγ and 1A mRNA are 345 and 277 bp, respectively.</p>
</caption>
<graphic alt-version="no" mime-subtype="gif" mimetype="image" position="float" xlink:href="S0007114507280584_fig3"></graphic>
</fig>
</p>
</sec>
<sec id="sec2-4">
<title>Effects of the peroxisome proliferator-activated receptor-γ antagonist bisphenol A diglycidyl ether on tumour necrosis factor-α expression in peripheral blood mononuclear cells and the phagocytic capacity of polymorphonuclear cells</title>
<p>To examine whether the activation of PPARγ by t10c12-CLA was associated with TNF-α production by PBMC and the phagocytic capacity of PMN, BADGE, a PPARγ antagonist, was added to the PBMC culture. The amount of PBMC TNF-α production and the stimulation of PMN phagocytic capacity by the culture supernatant fraction from PBMC treated with t10c12-CLA alone or in combination with BADGE (100 μ
<sc>m</sc>
) for 24 h were determined. As shown in
<xref ref-type="fig" rid="fig4">Fig. 4 (A)</xref>
, BADGE completely negated (
<italic>P</italic>
 < 0·001) the effect of t10c12-CLA on TNF-α production by PBMC when compared with the effect of the culture supernatant fraction without BADGE. The enhanced phagocytic capacity of PMN in response to the culture supernatant fraction from PBMC treated with t10c12-CLA (5–20 μ
<sc>m</sc>
) was inhibited (
<italic>P</italic>
 < 0·001) by the addition of BADGE to the PBMC culture (
<xref ref-type="fig" rid="fig4">Fig. 4 (B)</xref>
). RT-PCR analysis also showed that BADGE completely antagonised the TNF-α mRNA expression induced by t10c12-CLA in PBMC (
<xref ref-type="fig" rid="fig4">Fig. 4 (C)</xref>
).
<fig id="fig4" position="float">
<label>Fig. 4</label>
<caption>
<p>Effects of bisphenol A diglycidyl ether (BADGE), a PPARγ antagonist, on TNF-α expression in peripheral blood mononuclear cells (PBMC) and the phagocytic capacity of polymorphonuclear cells (PMN). (A) The amount of TNF-α in the culture supernatant fraction from PBMC (2 × 10
<sup>6</sup>
cells/ml) treated with either
<italic>trans</italic>
-10,
<italic>cis</italic>
-12-conjugated linoleic acid (t10c12-CLA) alone (
<private-char name="S0007114507280584_char1">
<inline-graphic mime-subtype="gif" mimetype="image" xlink:href="S0007114507280584_char1"></inline-graphic>
</private-char>
) or t10c12-CLA in combination with BADGE (□) at the indicated concentrations for 24 h was determined using ELISA. (B) The phagocytic capacity of PMN in response to the culture supernatant fractions from PBMC (2 × 10
<sup>6</sup>
cells/ml) treated with either t10c12-CLA alone or t10c12-CLA in combination with BADGE, at the indicated concentrations for 24 h. PMN (1 × 10
<sup>6</sup>
cells/ml per well) were incubated for 12 h. Cultures were supplemented with fluorescein isothiocyanate-latex beads for the final 1 h. Differences within the input groups of each t10c12-CLA concentration were analysed with Student's
<italic>t</italic>
test, separately. Data are means (
<italic>n</italic>
6), with standard deviations represented by vertical bars. * Mean value was significantly different from that for the culture supernatant fraction from PBMC treated with t10c12-CLA alone (
<italic>P</italic>
 < 0·05). (C) The effect of BADGE on TNF-α mRNA expression in porcine PBMC treated with t10c12-CLA for 3 h. RT-PCR analysis of TNF-α mRNA expression in PBMC treated with t10c12-CLA (a). Normalisation of TNF-α mRNA expression with 1A (b). The expected product sizes of TNF-α and 1A mRNA are 273 and 277 bp, respectively.</p>
</caption>
<graphic alt-version="no" mime-subtype="gif" mimetype="image" position="float" xlink:href="S0007114507280584_fig4"></graphic>
</fig>
</p>
</sec>
</sec>
<sec id="sec3" sec-type="discussion">
<title>Discussion</title>
<p>t10c12-CLA up regulated TNF-α production from porcine PBMC. The phagocytic capacity of porcine peripheral blood PMN was increased by the culture supernatant fraction from PBMC treated with t10c12-CLA. The phagocytic capacity of PMN was also increased by rpTNF-α dose-dependently. It was highest at the rpTNF-α concentration of 1 ng/ml and down regulated by 10 ng/ml. Similar to the phagocytic capacity kinetics of rpTNF-α, the rhTNF-α concentration of 1 ng/ml exhibited the highest capacity in porcine PMN phagocytosis, whereas 10 ng/ml showed low levels of PMN phagocytic capacity (Yang
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref52">2005</xref>
). It was, therefore, assumed that the dose of recombinant TNF-α of 1 ng/ml is the most effective dose responsible for the phagocytic capacity of porcine PMN. Anti-rpTNF-α pAb completely abolished the enhanced phagocytic capacity of porcine PMN induced by the culture supernatant fraction from PBMC treated with t10c12-CLA. These findings indicate that t10c12-CLA stimulates porcine PBMC to produce TNF-α, which enhances the phagocytic capacity of PMN. Several reports are in agreement with these findings. The phagocytic capacity of neutrophils has been reported to be up regulated by TNF-α (Shalaby
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref43">1985</xref>
). Dietary t10c12-CLA has been reported to increase TNF-α mRNA levels 12-fold in isolated adipocytes and non-adipocytes (Tsuboyama-Kasaoka
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref48">2000</xref>
). The up regulation of TNF-α by t10c12-CLA has also been observed in mouse splenocytes (Kelley
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref23">2002</xref>
). Up regulation of TNF-α mRNA induced by t10c12-CLA has numerous beneficial effects including anti-adipogenesis (Park
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref36">1999</xref>
; Tsuboyama-Kasaoka
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref48">2000</xref>
), anti-carcinogenesis (Ip
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref20">1999</xref>
), and immunostimulation (Kim
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref24">2003</xref>
). The TNF-α signal is transmitted via crosslinking of the membrane-bound receptor molecules, TNF receptor (TNF-R)α and TNF-Rβ, on target cells (Vandenabeele
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref49">1995</xref>
). After binding to its membrane-bound receptors, TNF-α mediates a wide range of effects such as the regulation of immune function, mediation of the inflammatory response and triggering of apoptosis (Hu,
<xref ref-type="bibr" rid="ref18">2003</xref>
).</p>
<p>It has been reported that CLA decreases the synthesis of prostaglandins, in particular PGE
<sub>2</sub>
(Li & Watkins,
<xref ref-type="bibr" rid="ref28">1998</xref>
), and PGE
<sub>2</sub>
has been shown to have profound down regulatory effects on various inflammatory cells (Levy
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref27">2001</xref>
; Harris
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref15">2002</xref>
). PGE
<sub>2</sub>
has been reported to regulate macrophage TNF-α production through negative feedback (Kunkel
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref25">1986</xref>
; Ikegami
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref19">2001</xref>
; Shinomiya
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref44">2001</xref>
). Therefore, one possible explanation for increased TNF-α expression in porcine PBMC treated with t10c12-CLA may be related to decreased PGE
<sub>2</sub>
regulation of TNF-α. On the other hand, this effect of CLA may be also a general effect of
<italic>n</italic>
-6 PUFA since
<italic>n</italic>
-6 PUFA are known to be pro-inflammatory (Toborek
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref47">1996</xref>
). It has been suggested that perturbation of PUFA metabolism by CLA will have an impact on eicosanoid formation and metabolism, closely linked to the biological activities of CLA (Banni
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref2">2004</xref>
).</p>
<p>In the present study, PPARγ expression in porcine PBMC was increased by t10c12-CLA treatment. This finding is in accord with other reports that CLA increased PPARγ expression in tissue and cells such as skeletal muscle (Meadus
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref29">2002</xref>
), adipocytes (Evans
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref13">2000</xref>
) and macrophages (Yu
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref54">2002</xref>
). When the binding affinity of t10c12-CLA to PPARγ was determined using a scintillation proximity assay, t10c12-CLA was found to be a ligand for PPARγ with an affinity ranging from 4·2 to 5·2 μ
<sc>m</sc>
(Belury
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref3">2002</xref>
). It was suggested that the ability of CLA to induce PPARγ-responsive genes may be due to direct binding of CLA to the PPARγ as well as the binding of active metabolites of CLA produced via Δ6 desaturase. Therefore, it was thought that the actions of t10c12-CLA may be associated with increased levels of PPARγ protein and/or activation of PPARγ by downstream metabolites.</p>
<p>It has been reported that PPAR activators slightly increase TNF-α production in Kupffer cells (Nakatani
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref33">2002</xref>
). Increased PPARγ mRNA expression in the mesenteric tissue could lead to mesenteric fat hypertrophy, which could actively participate through the synthesis of TNF-α (Desreumaux
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref12">1999</xref>
). These observations support the idea that the activation of PPARγ by t10c12-CLA can regulate TNF-α gene transcription in porcine PBMC. The present results reveal that t10c12-CLA stimulates both PPARγ and TNF-α expression in porcine PBMC. We used a PPARγ antagonist, BADGE, which is a PPARγ ligand with a K
<sub>d(app)</sub>
of 100 μ
<sc>m</sc>
(Wright
<italic>et al.</italic>
<xref ref-type="bibr" rid="ref50">2000</xref>
), to elucidate the role of PPARγ on TNF-α expression in porcine PBMC induced by t10c12-CLA. BADGE negated the effect of CLA on TNF-α expression. This BADGE-induced decrease in TNF-α production by PBMC diminished the enhancement of PMN phagocytic capacity induced by the t10c12-CLA-stimulated PBMC culture supernatant fraction. These results suggest that the effects of t10c12-CLA on TNF-α production in porcine PBMC may be dependent on the PPARγ pathway. Therefore, it can be concluded that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α production by PBMC via a PPARγ-dependent pathway.</p>
</sec>
</body>
<back>
<ack id="ack1">
<title>Acknowledgements</title>
<p>The present study was supported by the National R & D Programme Grant of The Ministry of Science and Technology (M1-0417-06-0005), National Livestock Research Institute and the Ministry of Education and Human Resources Development (MOE), the Ministry of Commerce, Industry and Energy (MOCIE) and the Ministry of Labour (MOLAB) through the fostering project of the Laboratory of Excellency. In addition, the authors appreciate the graduate fellowship provided by the Ministry of Education through the BK21 programme. The authors also thank Dr Daehyun Chung, Department of Information and Statistics, Chungbuk National University, Republic of Korea, for statistically analysing the data.</p>
</ack>
<ref-list>
<title>References</title>
<ref>
<citation citation-type="journal" id="ref1">
<name name-style="western">
<surname>Antal-Szalmás</surname>
<given-names>P</given-names>
</name>
,
<name name-style="western">
<surname>Poppelier</surname>
<given-names>MJ</given-names>
</name>
,
<name name-style="western">
<surname>Broekhuizen</surname>
<given-names>R</given-names>
</name>
,
<name name-style="western">
<surname>Verhoef</surname>
<given-names>J</given-names>
</name>
,
<name name-style="western">
<surname>van Strijp</surname>
<given-names>JA</given-names>
</name>
&
<name name-style="western">
<surname>van Kessel</surname>
<given-names>KP</given-names>
</name>
(
<year>2000</year>
)
<article-title>Diverging pathways for lipopolysaccharide and CD14 in human monocytes</article-title>
.
<source>Cytometry</source>
<volume>41</volume>
,
<fpage>279</fpage>
<lpage>288</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref2">
<name name-style="western">
<surname>Banni</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Petroni</surname>
<given-names>A</given-names>
</name>
,
<name name-style="western">
<surname>Blasevich</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Carta</surname>
<given-names>G</given-names>
</name>
,
<name name-style="western">
<surname>Cordeddu</surname>
<given-names>L</given-names>
</name>
,
<name name-style="western">
<surname>Murru</surname>
<given-names>E</given-names>
</name>
,
<name name-style="western">
<surname>Melis</surname>
<given-names>MP</given-names>
</name>
,
<name name-style="western">
<surname>Mahon</surname>
<given-names>A</given-names>
</name>
&
<name name-style="western">
<surname>Belury</surname>
<given-names>MA</given-names>
</name>
(
<year>2004</year>
)
<article-title>Conjugated linoleic acids (CLA) as precursors of a distinct family of PUFA</article-title>
.
<source>Lipids</source>
<volume>39</volume>
,
<fpage>1143</fpage>
<lpage>1146</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref3">
<name name-style="western">
<surname>Belury</surname>
<given-names>MA</given-names>
</name>
,
<name name-style="western">
<surname>Moya-Camarena</surname>
<given-names>SY</given-names>
</name>
,
<name name-style="western">
<surname>Lu</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Shi</surname>
<given-names>L</given-names>
</name>
,
<name name-style="western">
<surname>Leesnitzer</surname>
<given-names>LM</given-names>
</name>
&
<name name-style="western">
<surname>Blanchard</surname>
<given-names>SG</given-names>
</name>
(
<year>2002</year>
)
<article-title>Conjugated linoleic acid is an activator and ligand for peroxisome proliferator-activated receptor-gamma (PPARγ)</article-title>
.
<source>Nutr Res</source>
<volume>22</volume>
,
<fpage>817</fpage>
<lpage>824</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref4">
<name name-style="western">
<surname>Bishop-Bailey</surname>
<given-names>D</given-names>
</name>
&
<name name-style="western">
<surname>Wray</surname>
<given-names>J</given-names>
</name>
(
<year>2003</year>
)
<article-title>Peroxisome proliferator-activated receptors: a critical review on endogenous pathways for ligand generation</article-title>
.
<source>Prostaglandins Other Lipid Mediat</source>
<volume>71</volume>
,
<fpage>1</fpage>
<lpage>22</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref5">
<name name-style="western">
<surname>Braissant</surname>
<given-names>O</given-names>
</name>
,
<name name-style="western">
<surname>Foufelle</surname>
<given-names>F</given-names>
</name>
,
<name name-style="western">
<surname>Scotto</surname>
<given-names>C</given-names>
</name>
,
<name name-style="western">
<surname>Dauca</surname>
<given-names>M</given-names>
</name>
&
<name name-style="western">
<surname>Wahli</surname>
<given-names>W</given-names>
</name>
(
<year>1996</year>
)
<article-title>Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat</article-title>
.
<source>Endocrinology</source>
<volume>137</volume>
,
<fpage>354</fpage>
<lpage>366</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref6">
<name name-style="western">
<surname>Brown</surname>
<given-names>JM</given-names>
</name>
,
<name name-style="western">
<surname>Boysen</surname>
<given-names>MS</given-names>
</name>
,
<name name-style="western">
<surname>Jensen</surname>
<given-names>SS</given-names>
</name>
,
<name name-style="western">
<surname>Morrison</surname>
<given-names>RF</given-names>
</name>
,
<name name-style="western">
<surname>Storkson</surname>
<given-names>J</given-names>
</name>
,
<name name-style="western">
<surname>Lea-Currie</surname>
<given-names>R</given-names>
</name>
,
<name name-style="western">
<surname>Pariza</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Mandrup</surname>
<given-names>S</given-names>
</name>
&
<name name-style="western">
<surname>McIntosh</surname>
<given-names>MK</given-names>
</name>
(
<year>2003</year>
)
<article-title>Isomer-specific regulation of metabolism and PPARγ signaling by CLA in human preadipocytes</article-title>
.
<source>J Lipid Res</source>
<volume>44</volume>
,
<fpage>1287</fpage>
<lpage>1300</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref7">
<name name-style="western">
<surname>Calder</surname>
<given-names>PC</given-names>
</name>
(
<year>1996</year>
)
<article-title>Effects of fatty acids and dietary lipids on cells of the immune system</article-title>
.
<source>Proc Nutr Soc</source>
<volume>55</volume>
,
<fpage>127</fpage>
<lpage>150</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref8">
<name name-style="western">
<surname>Calder</surname>
<given-names>PC</given-names>
</name>
(
<year>1998</year>
)
<article-title>Dietary fatty acids and the immune system</article-title>
.
<source>Nutr Rev</source>
<volume>56</volume>
,
<fpage>S70</fpage>
<lpage>S83</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref9">
<name name-style="western">
<surname>Chawla</surname>
<given-names>A</given-names>
</name>
,
<name name-style="western">
<surname>Schwarz</surname>
<given-names>EJ</given-names>
</name>
,
<name name-style="western">
<surname>Dimaculangan</surname>
<given-names>DD</given-names>
</name>
&
<name name-style="western">
<surname>Lazar</surname>
<given-names>MA</given-names>
</name>
(
<year>1994</year>
)
<article-title>Peroxisome proliferator-activated receptor (PPAR) gamma: adipose-predominant expression and induction early in adipocyte differentiation</article-title>
.
<source>Endocrinology</source>
<volume>135</volume>
,
<fpage>798</fpage>
<lpage>800</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref10">
<name name-style="western">
<surname>Cunard</surname>
<given-names>R</given-names>
</name>
,
<name name-style="western">
<surname>Ricote</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>DiCampli</surname>
<given-names>D</given-names>
</name>
,
<name name-style="western">
<surname>Archer</surname>
<given-names>DC</given-names>
</name>
,
<name name-style="western">
<surname>Kahn</surname>
<given-names>DA</given-names>
</name>
,
<name name-style="western">
<surname>Glass</surname>
<given-names>CK</given-names>
</name>
&
<name name-style="western">
<surname>Kelly</surname>
<given-names>CJ</given-names>
</name>
(
<year>2002</year>
)
<article-title>Regulation of cytokine expression by ligands of peroxisome proliferators activated receptors</article-title>
.
<source>J Immunol</source>
<volume>168</volume>
,
<fpage>2795</fpage>
<lpage>2802</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref11">
<name name-style="western">
<surname>Daynes</surname>
<given-names>RA</given-names>
</name>
&
<name name-style="western">
<surname>Jones</surname>
<given-names>DC</given-names>
</name>
(
<year>2002</year>
)
<article-title>Emerging roles of PPARs in inflammation and immunity</article-title>
.
<source>Nat Rev Immunol</source>
<volume>2</volume>
,
<fpage>748</fpage>
<lpage>759</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref12">
<name name-style="western">
<surname>Desreumaux</surname>
<given-names>P</given-names>
</name>
,
<name name-style="western">
<surname>Ernst</surname>
<given-names>O</given-names>
</name>
,
<name name-style="western">
<surname>Geboes</surname>
<given-names>K</given-names>
</name>
,
<etal></etal>
(
<year>1999</year>
)
<article-title>Inflammatory alterations in mesenteric adipose tissue in Crohn's disease</article-title>
.
<source>Gastroenterology</source>
<volume>117</volume>
,
<fpage>73</fpage>
<lpage>81</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref13">
<name name-style="western">
<surname>Evans</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Pariza</surname>
<given-names>MW</given-names>
</name>
,
<name name-style="western">
<surname>Park</surname>
<given-names>Y</given-names>
</name>
,
<name name-style="western">
<surname>Curtis</surname>
<given-names>L</given-names>
</name>
,
<name name-style="western">
<surname>Kuebler</surname>
<given-names>B</given-names>
</name>
&
<name name-style="western">
<surname>McIntosh</surname>
<given-names>M</given-names>
</name>
(
<year>2000</year>
)
<article-title>
<italic>Trans</italic>
-10,
<italic>cis</italic>
-12 conjugated linoleic acid reduces triglyceride content while differentially affecting peroxisome proliferator activated receptor-γ2 and aP2 expression in 3T3-L1 adipocytes</article-title>
.
<source>Lipids</source>
<volume>36</volume>
,
<fpage>1223</fpage>
<lpage>1232</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref14">
<name name-style="western">
<surname>Granlund</surname>
<given-names>L</given-names>
</name>
,
<name name-style="western">
<surname>Juvet</surname>
<given-names>LK</given-names>
</name>
,
<name name-style="western">
<surname>Pederson</surname>
<given-names>JI</given-names>
</name>
&
<name name-style="western">
<surname>Nebb</surname>
<given-names>HI</given-names>
</name>
(
<year>2003</year>
)
<article-title>
<italic>Trans</italic>
10,
<italic>cis</italic>
12-conjugated linoleic acid prevents triacyglycerol accumulation in adipocytes by acting as a PPAR γ modulator</article-title>
.
<source>J Lipid Res</source>
<volume>44</volume>
,
<fpage>1441</fpage>
<lpage>1452</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref15">
<name name-style="western">
<surname>Harris</surname>
<given-names>SH</given-names>
</name>
,
<name name-style="western">
<surname>Padilla</surname>
<given-names>J</given-names>
</name>
,
<name name-style="western">
<surname>Koumas</surname>
<given-names>L</given-names>
</name>
,
<name name-style="western">
<surname>Ray</surname>
<given-names>D</given-names>
</name>
&
<name name-style="western">
<surname>Phipps</surname>
<given-names>RP</given-names>
</name>
(
<year>2002</year>
)
<article-title>Prostaglandins as modulators of immunity</article-title>
.
<source>Trends Immunol</source>
<volume>23</volume>
,
<fpage>144</fpage>
<lpage>150</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref16">
<name name-style="western">
<surname>Hed</surname>
<given-names>J</given-names>
</name>
,
<name name-style="western">
<surname>Hallden</surname>
<given-names>G</given-names>
</name>
,
<name name-style="western">
<surname>Johansson</surname>
<given-names>SG</given-names>
</name>
&
<name name-style="western">
<surname>Larsson</surname>
<given-names>P</given-names>
</name>
(
<year>1987</year>
)
<article-title>The use of fluorescence quenching in flow cytofluorometry to measure the attachment and ingestion phases in phagocytosis in peripheral blood without prior cell separation</article-title>
.
<source>J Immunol Methods</source>
<volume>101</volume>
,
<fpage>119</fpage>
<lpage>125</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref17">
<name name-style="western">
<surname>Houseknecht</surname>
<given-names>KL</given-names>
</name>
,
<name name-style="western">
<surname>Vanden Heuvel</surname>
<given-names>JP</given-names>
</name>
,
<name name-style="western">
<surname>Moya-Camarena</surname>
<given-names>SY</given-names>
</name>
,
<name name-style="western">
<surname>Portocarrero</surname>
<given-names>CP</given-names>
</name>
,
<name name-style="western">
<surname>Peck</surname>
<given-names>LW</given-names>
</name>
,
<name name-style="western">
<surname>Nickel</surname>
<given-names>KP</given-names>
</name>
&
<name name-style="western">
<surname>Belury</surname>
<given-names>MA</given-names>
</name>
(
<year>1998</year>
)
<article-title>Dietary conjugated linoleic acid normalizes impaired glucose tolerance in the Zucker diabetic fatty fa/fa rat</article-title>
.
<source>Biochem Biophys Res Commun.</source>
<volume>244</volume>
,
<fpage>678</fpage>
<lpage>682</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref18">
<name name-style="western">
<surname>Hu</surname>
<given-names>X</given-names>
</name>
(
<year>2003</year>
)
<article-title>Proteolytic signaling by TNFα: cascade activation and IκB degradation</article-title>
.
<source>Cytokine</source>
<volume>21</volume>
,
<fpage>286</fpage>
<lpage>294</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref19">
<name name-style="western">
<surname>Ikegami</surname>
<given-names>R</given-names>
</name>
,
<name name-style="western">
<surname>Sugimoto</surname>
<given-names>Y</given-names>
</name>
,
<name name-style="western">
<surname>Segi</surname>
<given-names>E</given-names>
</name>
,
<name name-style="western">
<surname>Katsuyama</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Karahashi</surname>
<given-names>H</given-names>
</name>
,
<name name-style="western">
<surname>Amano</surname>
<given-names>F</given-names>
</name>
,
<name name-style="western">
<surname>Maruyama</surname>
<given-names>T</given-names>
</name>
,
<name name-style="western">
<surname>Yamane</surname>
<given-names>H</given-names>
</name>
,
<name name-style="western">
<surname>Tsuchiya</surname>
<given-names>S</given-names>
</name>
&
<name name-style="western">
<surname>Ichikawa</surname>
<given-names>A</given-names>
</name>
(
<year>2001</year>
)
<article-title>The expression of prostaglandin E receptors EP
<sub>2</sub>
and EP
<sub>4</sub>
and their different regulation by lipopolysaccharide in C3H/HeN peritoneal macrophages</article-title>
.
<source>J Immunol</source>
<volume>166</volume>
,
<fpage>4689</fpage>
<lpage>4696</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref20">
<name name-style="western">
<surname>Ip</surname>
<given-names>MM</given-names>
</name>
,
<name name-style="western">
<surname>Masao-Welch</surname>
<given-names>PA</given-names>
</name>
,
<name name-style="western">
<surname>Shoemaker</surname>
<given-names>SF</given-names>
</name>
,
<name name-style="western">
<surname>Shea-Eaton</surname>
<given-names>WK</given-names>
</name>
&
<name name-style="western">
<surname>Ip</surname>
<given-names>C</given-names>
</name>
(
<year>1999</year>
)
<article-title>Conjugated linoleic acid inhibits proliferation and induces apoptosis of normal rat mammary epithelial cells in primary culture</article-title>
.
<source>Exp Cell Res</source>
<volume>250</volume>
,
<fpage>22</fpage>
<lpage>34</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref21">
<name name-style="western">
<surname>Jiang</surname>
<given-names>C</given-names>
</name>
,
<name name-style="western">
<surname>Ting</surname>
<given-names>AT</given-names>
</name>
&
<name name-style="western">
<surname>Seed</surname>
<given-names>B</given-names>
</name>
(
<year>1998</year>
)
<article-title>PPAR-γ agonists inhibit production of monocyte inflammatory cytokines</article-title>
.
<source>Nature</source>
<volume>391</volume>
,
<fpage>82</fpage>
<lpage>86</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref22">
<name name-style="western">
<surname>Kang</surname>
<given-names>JH</given-names>
</name>
,
<name name-style="western">
<surname>Kim</surname>
<given-names>JH</given-names>
</name>
,
<name name-style="western">
<surname>Chung</surname>
<given-names>CS</given-names>
</name>
,
<name name-style="western">
<surname>Lee</surname>
<given-names>CY</given-names>
</name>
&
<name name-style="western">
<surname>Yang</surname>
<given-names>MP</given-names>
</name>
(
<year>2004</year>
)
<article-title>Immunoenhancing effects of conjugated linoleic acid on phagocytic activity of porcine peripheral blood phagocytes</article-title>
.
<source>J Vet Clin</source>
<volume>21</volume>
,
<fpage>336</fpage>
<lpage>342</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref23">
<name name-style="western">
<surname>Kelley</surname>
<given-names>DS</given-names>
</name>
,
<name name-style="western">
<surname>Warren</surname>
<given-names>JM</given-names>
</name>
,
<name name-style="western">
<surname>Simon</surname>
<given-names>VA</given-names>
</name>
,
<name name-style="western">
<surname>Bartolini</surname>
<given-names>G</given-names>
</name>
,
<name name-style="western">
<surname>Mackey</surname>
<given-names>BE</given-names>
</name>
&
<name name-style="western">
<surname>Erickson</surname>
<given-names>KL</given-names>
</name>
(
<year>2002</year>
)
<article-title>Similar effects of c9, t11-CLA and t10, c12-CLA on immune cell functions in mice</article-title>
.
<source>Lipids</source>
<volume>37</volume>
,
<fpage>725</fpage>
<lpage>728</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref24">
<name name-style="western">
<surname>Kim</surname>
<given-names>JH</given-names>
</name>
,
<name name-style="western">
<surname>Chung</surname>
<given-names>CS</given-names>
</name>
,
<name name-style="western">
<surname>Lee</surname>
<given-names>CY</given-names>
</name>
&
<name name-style="western">
<surname>Yang</surname>
<given-names>MP</given-names>
</name>
(
<year>2003</year>
)
<article-title>Immunoenhancing effects of conjugated linoleic acid on chemotactic activity of porcine blood polymorphonuclear cells</article-title>
.
<source>J Vet Clin</source>
<volume>20</volume>
,
<fpage>1</fpage>
<lpage>6</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref25">
<name name-style="western">
<surname>Kunkel</surname>
<given-names>SL</given-names>
</name>
,
<name name-style="western">
<surname>Wiggins</surname>
<given-names>RC</given-names>
</name>
,
<name name-style="western">
<surname>Chensue</surname>
<given-names>SW</given-names>
</name>
&
<name name-style="western">
<surname>Larrick</surname>
<given-names>J</given-names>
</name>
(
<year>1986</year>
)
<article-title>Regulation of macrophage tumor necrosis factor production by prostaglandin E
<sub>2</sub>
</article-title>
.
<source>Biochem Biophys Res Commun</source>
<volume>137</volume>
,
<fpage>404</fpage>
<lpage>410</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref26">
<name name-style="western">
<surname>Lee</surname>
<given-names>KN</given-names>
</name>
,
<name name-style="western">
<surname>Kritchevsky</surname>
<given-names>D</given-names>
</name>
&
<name name-style="western">
<surname>Pariza</surname>
<given-names>MW</given-names>
</name>
(
<year>1994</year>
)
<article-title>Conjugated linoleic acid and atherosclerosis in rabbits</article-title>
.
<source>Atherosclerosis</source>
<volume>108</volume>
,
<fpage>19</fpage>
<lpage>25</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref27">
<name name-style="western">
<surname>Levy</surname>
<given-names>BD</given-names>
</name>
,
<name name-style="western">
<surname>Clish</surname>
<given-names>CB</given-names>
</name>
,
<name name-style="western">
<surname>Schmidt</surname>
<given-names>B</given-names>
</name>
,
<name name-style="western">
<surname>Gronert</surname>
<given-names>K</given-names>
</name>
&
<name name-style="western">
<surname>Serhan</surname>
<given-names>CN</given-names>
</name>
(
<year>2001</year>
)
<article-title>Lipid mediator class switching during acute inflammation: signals in resolution</article-title>
.
<source>Nat Immunol</source>
<volume>2</volume>
,
<fpage>612</fpage>
<lpage>619</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref28">
<name name-style="western">
<surname>Li</surname>
<given-names>Y</given-names>
</name>
&
<name name-style="western">
<surname>Watkins</surname>
<given-names>BA</given-names>
</name>
(
<year>1998</year>
)
<article-title>Conjugated linoleic acids alter bone fatty acid composition and reduce
<italic>ex vivo</italic>
prostaglandin E2 biosynthesis in rats fed
<italic>n</italic>
-6 or
<italic>n</italic>
-3 fatty acids</article-title>
.
<source>Lipids</source>
<volume>33</volume>
,
<fpage>417</fpage>
<lpage>425</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref29">
<name name-style="western">
<surname>Meadus</surname>
<given-names>WJ</given-names>
</name>
,
<name name-style="western">
<surname>Maclnnis</surname>
<given-names>R</given-names>
</name>
&
<name name-style="western">
<surname>Dugan</surname>
<given-names>MER</given-names>
</name>
(
<year>2002</year>
)
<article-title>Prolonged dietary treatment with conjugated linoleic acid stimulates porcine muscle peroxisome proliferator activated receptor γ and glutamine-fructose aminotransferase gene expression
<italic>in vivo</italic>
</article-title>
.
<source>J Mol Endocrinol</source>
<volume>38</volume>
,
<fpage>79</fpage>
<lpage>86</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref30">
<name name-style="western">
<surname>Mexmain</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Gualde</surname>
<given-names>N</given-names>
</name>
,
<name name-style="western">
<surname>Aldigier</surname>
<given-names>JC</given-names>
</name>
,
<name name-style="western">
<surname>Motta</surname>
<given-names>C</given-names>
</name>
,
<name name-style="western">
<surname>Chable-Rabinovitch</surname>
<given-names>H</given-names>
</name>
&
<name name-style="western">
<surname>Rigaud</surname>
<given-names>M</given-names>
</name>
(
<year>1984</year>
)
<article-title>Specific binding of 15 HETE to lymphocyte. Effects on the fluidity of plasmatic membranes</article-title>
.
<source>Prostaglandins Leukot Med</source>
<volume>13</volume>
,
<fpage>93</fpage>
<lpage>97</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref31">
<name name-style="western">
<surname>Miles</surname>
<given-names>EA</given-names>
</name>
,
<name name-style="western">
<surname>Allen</surname>
<given-names>E</given-names>
</name>
&
<name name-style="western">
<surname>Calder</surname>
<given-names>PC</given-names>
</name>
(
<year>2002</year>
)
<article-title>
<italic>In vitro</italic>
effects of eicosanoids derived from different 20-carbon fatty acids on production of monocytic-derived cytokines in human whole blood cultures</article-title>
.
<source>Cytokine</source>
<volume>20</volume>
,
<fpage>215</fpage>
<lpage>223</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref32">
<name name-style="western">
<surname>Moya-Camarena</surname>
<given-names>SY</given-names>
</name>
,
<name name-style="western">
<surname>Vanden Heuvel</surname>
<given-names>JP</given-names>
</name>
,
<name name-style="western">
<surname>Blanchard</surname>
<given-names>SG</given-names>
</name>
,
<name name-style="western">
<surname>Leesnitzer</surname>
<given-names>LA</given-names>
</name>
&
<name name-style="western">
<surname>Belury</surname>
<given-names>MA</given-names>
</name>
(
<year>1999</year>
)
<article-title>Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARα</article-title>
.
<source>J Lipid Res.</source>
<volume>40</volume>
,
<fpage>1426</fpage>
<lpage>1433</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref33">
<name name-style="western">
<surname>Nakatani</surname>
<given-names>T</given-names>
</name>
,
<name name-style="western">
<surname>Tsuboyama-Kasaoka</surname>
<given-names>N</given-names>
</name>
,
<name name-style="western">
<surname>Takahashi</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Miura</surname>
<given-names>S</given-names>
</name>
&
<name name-style="western">
<surname>Ezaki</surname>
<given-names>O</given-names>
</name>
(
<year>2002</year>
)
<article-title>Mechanism for peroxisome proliferator-activated receptor-α activator-induced up-regulation of UCP2 mRNA in rodent hepatocytes</article-title>
.
<source>J Biol Chem</source>
<volume>277</volume>
,
<fpage>9562</fpage>
<lpage>9569</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref34">
<name name-style="western">
<surname>Palombo</surname>
<given-names>JD</given-names>
</name>
,
<name name-style="western">
<surname>Gangguly</surname>
<given-names>A</given-names>
</name>
,
<name name-style="western">
<surname>Bistran</surname>
<given-names>BR</given-names>
</name>
&
<name name-style="western">
<surname>Menard</surname>
<given-names>MP</given-names>
</name>
(
<year>2002</year>
)
<article-title>The antiproliferative effects of biologically active isomers of conjugated linoleic acid on human colorectal and prostatic cancer cells</article-title>
.
<source>Cancer Lett</source>
<volume>177</volume>
,
<fpage>163</fpage>
<lpage>172</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref35">
<name name-style="western">
<surname>Pariza</surname>
<given-names>MW</given-names>
</name>
,
<name name-style="western">
<surname>Park</surname>
<given-names>Y</given-names>
</name>
&
<name name-style="western">
<surname>Cook</surname>
<given-names>ME</given-names>
</name>
(
<year>2001</year>
)
<article-title>The biologically active isomers of conjugated linoleic acid</article-title>
.
<source>Prog Lipid Res</source>
<volume>40</volume>
,
<fpage>283</fpage>
<lpage>298</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref36">
<name name-style="western">
<surname>Park</surname>
<given-names>Y</given-names>
</name>
,
<name name-style="western">
<surname>Storkson</surname>
<given-names>JM</given-names>
</name>
,
<name name-style="western">
<surname>Albright</surname>
<given-names>KJ</given-names>
</name>
,
<name name-style="western">
<surname>Liu</surname>
<given-names>W</given-names>
</name>
&
<name name-style="western">
<surname>Pariza</surname>
<given-names>MW</given-names>
</name>
(
<year>1999</year>
)
<article-title>Evidence that the
<italic>trans</italic>
-10,
<italic>cis</italic>
-12 isomer of conjugated linoleic acid induces body composition changes in mice</article-title>
.
<source>Lipids</source>
<volume>34</volume>
,
<fpage>235</fpage>
<lpage>241</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref37">
<name name-style="western">
<surname>Rajakangas</surname>
<given-names>J</given-names>
</name>
,
<name name-style="western">
<surname>Basu</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Salminen</surname>
<given-names>I</given-names>
</name>
&
<name name-style="western">
<surname>Mutanen</surname>
<given-names>M</given-names>
</name>
(
<year>2003</year>
)
<article-title>Adenoma growth stimulation by the
<italic>trans</italic>
-10,
<italic>cis</italic>
-12 isomer of conjugated linoleic acid (CLA) is associated with changes in mucosal NF-κB and cyclin D1 protein levels in the Min mouse</article-title>
.
<source>J Nutr</source>
<volume>133</volume>
,
<fpage>1943</fpage>
<lpage>1948</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref38">
<name name-style="western">
<surname>Ricote</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Li</surname>
<given-names>AA</given-names>
</name>
,
<name name-style="western">
<surname>Willsons</surname>
<given-names>TM</given-names>
</name>
,
<name name-style="western">
<surname>Kelly</surname>
<given-names>CJ</given-names>
</name>
&
<name name-style="western">
<surname>Glass</surname>
<given-names>CK</given-names>
</name>
(
<year>1998</year>
)
<article-title>The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation</article-title>
.
<source>Nature</source>
<volume>391</volume>
,
<fpage>79</fpage>
<lpage>82</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref39">
<name name-style="western">
<surname>Risérus</surname>
<given-names>U</given-names>
</name>
,
<name name-style="western">
<surname>Smedman</surname>
<given-names>A</given-names>
</name>
,
<name name-style="western">
<surname>Basu</surname>
<given-names>S</given-names>
</name>
&
<name name-style="western">
<surname>Vessby</surname>
<given-names>B</given-names>
</name>
(
<year>2004</year>
)
<article-title>Metabolic effects of conjugated linoleic acid in humans: the Swedish experience</article-title>
.
<source>Am J Clin Nutr</source>
<volume>79</volume>
,
<fpage>1146S</fpage>
<lpage>1148S</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref40">
<name name-style="western">
<surname>Ryder</surname>
<given-names>JW</given-names>
</name>
,
<name name-style="western">
<surname>Portocarrero</surname>
<given-names>CP</given-names>
</name>
,
<name name-style="western">
<surname>Song</surname>
<given-names>XM</given-names>
</name>
,
<etal></etal>
(
<year>2001</year>
)
<article-title>Isomer-specific antidiabetic properties of conjugated linoleic acid. Improved glucose tolerance, skeletal muscle insulin action, and UCP-2 gene expression</article-title>
.
<source>Diabetes</source>
<volume>50</volume>
,
<fpage>1149</fpage>
<lpage>1157</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref41">
<name name-style="western">
<surname>Schoonjans</surname>
<given-names>K</given-names>
</name>
,
<name name-style="western">
<surname>Martin</surname>
<given-names>G</given-names>
</name>
,
<name name-style="western">
<surname>Staels</surname>
<given-names>B</given-names>
</name>
&
<name name-style="western">
<surname>Auwerx</surname>
<given-names>J</given-names>
</name>
(
<year>1997</year>
)
<article-title>Peroxisome proliferator-activated receptors, orphans with ligands and functions</article-title>
.
<source>Curr Opin Lipidol</source>
<volume>8</volume>
,
<fpage>159</fpage>
<lpage>166</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref42">
<name name-style="western">
<surname>Schoonjans</surname>
<given-names>K</given-names>
</name>
,
<name name-style="western">
<surname>Staels</surname>
<given-names>B</given-names>
</name>
&
<name name-style="western">
<surname>Auwerx</surname>
<given-names>J</given-names>
</name>
(
<year>1996</year>
)
<article-title>Role of the peroxisome proliferator-activated receptor (PPAR) in mediating the effects of fibrates and fatty acids on gene expression</article-title>
.
<source>J Lipid Res</source>
<volume>37</volume>
,
<fpage>907</fpage>
<lpage>925</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref43">
<name name-style="western">
<surname>Shalaby</surname>
<given-names>MR</given-names>
</name>
,
<name name-style="western">
<surname>Aggarwal</surname>
<given-names>BB</given-names>
</name>
,
<name name-style="western">
<surname>Rinderknecht</surname>
<given-names>E</given-names>
</name>
,
<name name-style="western">
<surname>Svedersky</surname>
<given-names>LP</given-names>
</name>
,
<name name-style="western">
<surname>Finkle</surname>
<given-names>BS</given-names>
</name>
&
<name name-style="western">
<surname>Palladino</surname>
<given-names>MA</given-names>
</name>
(
<year>1985</year>
)
<article-title>Activation of human polymorphonuclear neutrophil functions by interferon-γ and tumor necrosis factors</article-title>
.
<source>J Immunol</source>
<volume>135</volume>
,
<fpage>2069</fpage>
<lpage>2073</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref44">
<name name-style="western">
<surname>Shinomiya</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Naraba</surname>
<given-names>H</given-names>
</name>
,
<name name-style="western">
<surname>Ueno</surname>
<given-names>A</given-names>
</name>
,
<etal></etal>
(
<year>2001</year>
)
<article-title>Regulation of TNFα and interleukin-10 production by prostaglandin I(2) and E(2): studies with prostaglandin receptor-deficient mice and prostaglandin E-receptor subtype-selective synthetic agonists</article-title>
.
<source>Biochem Pharmacol</source>
<volume>61</volume>
,
<fpage>1153</fpage>
<lpage>1160</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref45">
<name name-style="western">
<surname>Smedman</surname>
<given-names>A</given-names>
</name>
,
<name name-style="western">
<surname>Basu</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Jovinge</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Fredrikson</surname>
<given-names>GN</given-names>
</name>
&
<name name-style="western">
<surname>Vessby</surname>
<given-names>B</given-names>
</name>
(
<year>2005</year>
)
<article-title>Conjugated linoleic acid increased C-reactive protein in human subjects</article-title>
.
<source>Br J Nutr</source>
<volume>94</volume>
,
<fpage>791</fpage>
<lpage>795</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref46">
<name name-style="western">
<surname>Smedman</surname>
<given-names>A</given-names>
</name>
&
<name name-style="western">
<surname>Vessby</surname>
<given-names>B</given-names>
</name>
(
<year>2001</year>
)
<article-title>Conjugated linoleic acid supplementation in humans – metabolic effects</article-title>
.
<source>J Nutr</source>
<volume>36</volume>
,
<fpage>773</fpage>
<lpage>781</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref47">
<name name-style="western">
<surname>Toborek</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Barger</surname>
<given-names>SW</given-names>
</name>
,
<name name-style="western">
<surname>Mattson</surname>
<given-names>MP</given-names>
</name>
,
<name name-style="western">
<surname>Barve</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>McClain</surname>
<given-names>CJ</given-names>
</name>
&
<name name-style="western">
<surname>Hennig</surname>
<given-names>B</given-names>
</name>
(
<year>1996</year>
)
<article-title>Linoleic acid and TNF-α cross-amplify oxidative injury and dysfunction of endothelial cell</article-title>
.
<source>J Lipid Res</source>
<volume>37</volume>
,
<fpage>123</fpage>
<lpage>135</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref48">
<name name-style="western">
<surname>Tsuboyama-Kasaoka</surname>
<given-names>N</given-names>
</name>
,
<name name-style="western">
<surname>Takahashi</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Tanemura</surname>
<given-names>K</given-names>
</name>
,
<name name-style="western">
<surname>Kim</surname>
<given-names>HJ</given-names>
</name>
,
<name name-style="western">
<surname>Tange</surname>
<given-names>T</given-names>
</name>
,
<name name-style="western">
<surname>Okuyama</surname>
<given-names>H</given-names>
</name>
,
<name name-style="western">
<surname>Kasai</surname>
<given-names>M</given-names>
</name>
,
<name name-style="western">
<surname>Ikemoto</surname>
<given-names>S</given-names>
</name>
&
<name name-style="western">
<surname>Ezaki</surname>
<given-names>O</given-names>
</name>
(
<year>2000</year>
)
<article-title>Conjugated linoleic acid supplementation reduces adipose tissue by apoptosis and develops lipodystrophy in mice</article-title>
.
<source>Diabetes</source>
<volume>49</volume>
,
<fpage>1534</fpage>
<lpage>1542</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref49">
<name name-style="western">
<surname>Vandenabeele</surname>
<given-names>P</given-names>
</name>
,
<name name-style="western">
<surname>Declercq</surname>
<given-names>W</given-names>
</name>
,
<name name-style="western">
<surname>Beyaert</surname>
<given-names>R</given-names>
</name>
&
<name name-style="western">
<surname>Fiers</surname>
<given-names>W</given-names>
</name>
(
<year>1995</year>
)
<article-title>Two tumor necrosis factor receptors: structures and function</article-title>
.
<source>Trends Cell Biol</source>
<volume>5</volume>
,
<fpage>392</fpage>
<lpage>399</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref50">
<name name-style="western">
<surname>Wright</surname>
<given-names>HM</given-names>
</name>
,
<name name-style="western">
<surname>Clish</surname>
<given-names>CB</given-names>
</name>
,
<name name-style="western">
<surname>Mikami</surname>
<given-names>T</given-names>
</name>
,
<name name-style="western">
<surname>Hauser</surname>
<given-names>S</given-names>
</name>
,
<name name-style="western">
<surname>Yanagi</surname>
<given-names>K</given-names>
</name>
,
<name name-style="western">
<surname>Hiramatsu</surname>
<given-names>R</given-names>
</name>
,
<name name-style="western">
<surname>Serhan</surname>
<given-names>CN</given-names>
</name>
&
<name name-style="western">
<surname>Spiegelman</surname>
<given-names>BM</given-names>
</name>
(
<year>2000</year>
)
<article-title>A synthetic antagonist for the peroxisome proliferator-activated receptor γ inhibits adipocyte differentiation</article-title>
.
<source>J Biol Chem</source>
<volume>275</volume>
,
<fpage>1873</fpage>
<lpage>1877</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref51">
<name name-style="western">
<surname>Xu</surname>
<given-names>HE</given-names>
</name>
,
<name name-style="western">
<surname>Lambert</surname>
<given-names>MH</given-names>
</name>
,
<name name-style="western">
<surname>Montana</surname>
<given-names>VG</given-names>
</name>
,
<etal></etal>
(
<year>1999</year>
)
<article-title>Molecular recognition of fatty acids by peroxisome proliferator-activated receptors</article-title>
.
<source>Mol Cell</source>
<volume>3</volume>
,
<fpage>397</fpage>
<lpage>403</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref52">
<name name-style="western">
<surname>Yang</surname>
<given-names>MP</given-names>
</name>
,
<name name-style="western">
<surname>Ko</surname>
<given-names>IK</given-names>
</name>
,
<name name-style="western">
<surname>Kang</surname>
<given-names>JH</given-names>
</name>
,
<name name-style="western">
<surname>Song</surname>
<given-names>DH</given-names>
</name>
,
<name name-style="western">
<surname>Lee</surname>
<given-names>GS</given-names>
</name>
&
<name name-style="western">
<surname>Jeung</surname>
<given-names>EB</given-names>
</name>
(
<year>2005</year>
)
<article-title>Egg white derivatives induce tumor necrosis factor-alpha expression in porcine peripheral blood mononuclear cells</article-title>
.
<source>Vet Immunol Immunopathol</source>
<volume>106</volume>
,
<fpage>129</fpage>
<lpage>138</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref53">
<name name-style="western">
<surname>Yang</surname>
<given-names>MP</given-names>
</name>
,
<name name-style="western">
<surname>Lee</surname>
<given-names>KJ</given-names>
</name>
,
<name name-style="western">
<surname>Yun</surname>
<given-names>SM</given-names>
</name>
,
<name name-style="western">
<surname>Kim</surname>
<given-names>JH</given-names>
</name>
,
<name name-style="western">
<surname>Ko</surname>
<given-names>IK</given-names>
</name>
&
<name name-style="western">
<surname>Jeung</surname>
<given-names>EB</given-names>
</name>
(
<year>2002</year>
)
<article-title>Feline interleukin-8 expression in peripheral blood mononuclear cells induced by egg white derivatives</article-title>
.
<source>Vet Immunol Immunopathol</source>
<volume>86</volume>
,
<fpage>43</fpage>
<lpage>53</lpage>
</citation>
</ref>
<ref>
<citation citation-type="journal" id="ref54">
<name name-style="western">
<surname>Yu</surname>
<given-names>Y</given-names>
</name>
,
<name name-style="western">
<surname>Correll</surname>
<given-names>PH</given-names>
</name>
&
<name name-style="western">
<surname>Vanden Heuvel</surname>
<given-names>JP</given-names>
</name>
(
<year>2002</year>
)
<article-title>Conjugated linoleic acid decreases production of proinflammatory products in macrophages: evidence for a PPARγ-dependent mechanism</article-title>
.
<source>Biochim Biophys Acta</source>
<volume>1581</volume>
,
<fpage>89</fpage>
<lpage>99</lpage>
</citation>
</ref>
</ref-list>
</back>
</article>
</istex:document>
</istex:metadataXml>
<mods version="3.6">
<titleInfo lang="en">
<title>Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
</titleInfo>
<titleInfo type="alternative" lang="en" contentType="CDATA">
<title>Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro</title>
</titleInfo>
<name type="personal">
<namePart type="given">Ji-Houn</namePart>
<namePart type="family">Kang</namePart>
<affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
<role>
<roleTerm type="text">author</roleTerm>
</role>
</name>
<name type="personal">
<namePart type="given">Geun-Shik</namePart>
<namePart type="family">Lee</namePart>
<affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
<role>
<roleTerm type="text">author</roleTerm>
</role>
</name>
<name type="personal">
<namePart type="given">Eui-Bae</namePart>
<namePart type="family">Jeung</namePart>
<affiliation>Laboratory of Veterinary Biochemistry and Molecular Biology, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
<role>
<roleTerm type="text">author</roleTerm>
</role>
</name>
<name type="personal" displayLabel="corresp">
<namePart type="given">Mhan-Pyo</namePart>
<namePart type="family">Yang</namePart>
<affiliation>Laboratory of Veterinary Internal Medicine, Department of Veterinary Medicine, College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea</affiliation>
<affiliation>E-mail: mpyang@chungbuk.ac.kr</affiliation>
<role>
<roleTerm type="text">author</roleTerm>
</role>
</name>
<typeOfResource>text</typeOfResource>
<genre type="research-article" displayLabel="research-article" authority="ISTEX" authorityURI="https://content-type.data.istex.fr" valueURI="https://content-type.data.istex.fr/ark:/67375/XTP-1JC4F85T-7">research-article</genre>
<originInfo>
<publisher>Cambridge University Press</publisher>
<place>
<placeTerm type="text">Cambridge, UK</placeTerm>
</place>
<dateIssued encoding="w3cdtf">2007</dateIssued>
<copyrightDate encoding="w3cdtf">2007</copyrightDate>
</originInfo>
<language>
<languageTerm type="code" authority="iso639-2b">eng</languageTerm>
<languageTerm type="code" authority="rfc3066">en</languageTerm>
</language>
<abstract type="normal" lang="en">Trans-10, cis-12-conjugated linoleic acid (t10c12-CLA) has been shown to alter immune function. PPARγ has been shown to potentially play an important role in regulating inflammatory and immune responses by modulating the activity of monocytes and macrophages. Previous studies have indicated that the phagocytic capacity of porcine peripheral blood polymorphonuclear cells (PMN) was enhanced by the culture supernatant fraction from t10c12-CLA-stimulated porcine peripheral blood mononuclear cells (PBMC) but not by t10c12-CLA itself. In the present study, we examined the effects of t10c12-CLA on PPARγ and TNF-α expression of porcine PBMC and the phagocytic capacity of PMN. t10c12-CLA increased TNF-α mRNA expression and production by PBMC. The phagocytic capacity of porcine PMN was enhanced by either culture supernatant fraction from PBMC treated with t10c12-CLA or recombinant porcine (rp) TNF-α. Anti-rpTNF-α polyclonal antibody inhibited the enhancement of PMN phagocytic capacity. t10c12-CLA also up regulated PPARγ mRNA expression in porcine PBMC. Bisphenol A diglycidyl ether, a PPARγ antagonist, not only completely negated the t10c12-CLA-stimulating effects on TNF-α expression and production by porcine PBMC, but also decreased the enhancement of PMN phagocytic capacity by the t10c12-CLA-stimulated porcine PBMC culture supernatant fraction. These results suggest that t10c12-CLA has an immunostimulating effect on porcine PMN phagocytic capacity, which is mediated by TNF-α from PBMC via a PPARγ-dependent pathway.</abstract>
<note>Abbreviations: BADGE, bisphenol A diglycidyl ether; CLA, conjugated linoleic acid; FITC, fluorescein isothiocyanate; PAb, polyclonal antibody; PBMC, peripheral blood mononuclear cells; PMN, polymorphonuclear cells; rh, recombinant human; rp, recombinant porcine; t10c12, trans-10, cis-12</note>
<subject lang="en">
<genre></genre>
<topic>Conjugated linoleic acid</topic>
<topic>Peroxisome proliferator-activated receptor γ</topic>
<topic>Tumour necrosis factor-α</topic>
<topic>Phagocytosis</topic>
<topic>Neutrophils</topic>
</subject>
<relatedItem type="host">
<titleInfo>
<title>British Journal of Nutrition</title>
</titleInfo>
<titleInfo type="abbreviated">
<title>Br J Nutr</title>
</titleInfo>
<genre type="journal" authority="ISTEX" authorityURI="https://publication-type.data.istex.fr" valueURI="https://publication-type.data.istex.fr/ark:/67375/JMC-0GLKJH51-B">journal</genre>
<identifier type="ISSN">0007-1145</identifier>
<identifier type="eISSN">1475-2662</identifier>
<identifier type="PublisherID">BJN</identifier>
<part>
<date>2007</date>
<detail type="volume">
<caption>vol.</caption>
<number>97</number>
</detail>
<detail type="issue">
<caption>no.</caption>
<number>1</number>
</detail>
<extent unit="pages">
<start>117</start>
<end>125</end>
<total>9</total>
</extent>
</part>
</relatedItem>
<identifier type="istex">3865D293C133D8D879BCDFF7CB068779E57E9D1C</identifier>
<identifier type="ark">ark:/67375/6GQ-7SFQ1NP1-J</identifier>
<identifier type="DOI">10.1017/S0007114507280584</identifier>
<identifier type="PII">S0007114507280584</identifier>
<identifier type="ArticleID">01992</identifier>
<accessCondition type="use and reproduction" contentType="copyright">Copyright © The Authors 2007</accessCondition>
<recordInfo>
<recordContentSource authority="ISTEX" authorityURI="https://loaded-corpus.data.istex.fr" valueURI="https://loaded-corpus.data.istex.fr/ark:/67375/XBH-G3RCRD03-V">cambridge</recordContentSource>
<recordOrigin>Copyright © The Authors 2007</recordOrigin>
</recordInfo>
</mods>
<json:item>
<extension>json</extension>
<original>false</original>
<mimetype>application/json</mimetype>
<uri>https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/record.json</uri>
</json:item>
</metadata>
<annexes>
<json:item>
<extension>gif</extension>
<original>true</original>
<mimetype>image/gif</mimetype>
<uri>https://api.istex.fr/ark:/67375/6GQ-7SFQ1NP1-J/annexes.gif</uri>
</json:item>
</annexes>
<serie></serie>
</istex>
</record>

Pour manipuler ce document sous Unix (Dilib)

EXPLOR_STEP=$WICRI_ROOT/Sante/explor/MersV1/Data/Istex/Corpus
HfdSelect -h $EXPLOR_STEP/biblio.hfd -nk 001305 | SxmlIndent | more

Ou

HfdSelect -h $EXPLOR_AREA/Data/Istex/Corpus/biblio.hfd -nk 001305 | SxmlIndent | more

Pour mettre un lien sur cette page dans le réseau Wicri

{{Explor lien
   |wiki=    Sante
   |area=    MersV1
   |flux=    Istex
   |étape=   Corpus
   |type=    RBID
   |clé=     ISTEX:3865D293C133D8D879BCDFF7CB068779E57E9D1C
   |texte=   Trans-10, cis-12-conjugated linoleic acid increases phagocytosis of porcine peripheral blood polymorphonuclear cells in vitro
}}

Wicri

This area was generated with Dilib version V0.6.33.
Data generation: Mon Apr 20 23:26:43 2020. Site generation: Sat Mar 27 09:06:09 2021