Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
2.
Histopathology ; 52(3): 348-54, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18269586

ABSTRACT

AIMS: Neural (N)-cadherin belongs to a group of transmembrane molecules with a crucial role in tissue morphogenesis and maintenance of an epithelioid phenotype and increased N-cadherin expression is implicated in tumour progression and dedifferentiation. The aim was to determine whether evaluation of N-cadherin in pulmonary tumours might assist in identifying lesions with more aggressive potential. METHODS AND RESULTS: One hundred and fifty-five pulmonary lesions were analysed for N-cadherin expression using immunohistochemistry, including neuroendocrine hyperplasia (n = 3), typical carcinoid (n = 59), atypical carcinoid (n = 12), small cell lung carcinoma (n = 11), large cell neuroendocrine carcinoma (n = 12), adenocarcinoma (n = 35) and squamous cell carcinoma (n = 23). Lymph node status was correlated with immunohistochemical expression. N-cadherin expression was demonstrated in all cases of neuroendocrine hyperplasia, 96% of typical carcinoids, 83% of atypical carcinoids, 63% of the small cell lung carcinomas and 32% of large cell neuroendocrine carcinomas. Over 90% of the adenocarcinomas and 100% of the squamous cell carcinomas were negative. Increased N-cadherin expression in typical carcinoids was associated with negative lymph node status (P < 0.001). DISCUSSION: N-cadherin is differentially expressed in pulmonary tumours and is predominantly observed in neuroendocrine lung lesions with high expression in typical and atypical pulmonary carcinoids. The level of expression of N-cadherin between types of lung tumours does not appear to indicate malignant potential or aggressive behaviour.


Subject(s)
Adenocarcinoma/metabolism , Cadherins/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Biomarkers, Tumor/metabolism , Carcinoid Tumor/metabolism , Carcinoid Tumor/secondary , Carcinoid Tumor/surgery , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/surgery , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/secondary , Carcinoma, Small Cell/surgery , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Fluorescent Antibody Technique, Direct , Humans , Hyperplasia , Immunoenzyme Techniques , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology
3.
Transpl Infect Dis ; 6(2): 77-80, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15522109

ABSTRACT

This report describes the clinical course of a liver transplant recipient in whom cavitary pneumonia developed due to Legionella pneumophila. We review the experience with cavitary pulmonary processes caused by Legionella species in liver allograft recipients and describe the diagnostic microbiology of this organism. The clinical course of this patient demonstrates the importance of considering legionellosis in the differential diagnosis of lung abscesses after liver transplantation and the diagnostic difficulties encountered with this bacterium.


Subject(s)
Legionella pneumophila/isolation & purification , Legionnaires' Disease/microbiology , Liver Transplantation/adverse effects , Lung Abscess/microbiology , Adult , Female , Humans , Legionnaires' Disease/diagnosis , Lung Abscess/diagnosis , Transplantation, Homologous/adverse effects
4.
Gene Ther ; 10(18): 1608-15, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12907953

ABSTRACT

To increase the levels of pulmonary gene transfer by nonviral vectors, we have adopted electroporation protocols for use in the lung. A volume of 100-200 microl of purified plasmid DNA suspended in saline was instilled into the lungs of anesthetized mice. Plasmids expressed luciferase, or beta-galactosidase under control of the CMV immediate-early promoter and enhancer. Immediately following delivery, a series of eight square wave electric pulses of 10 ms duration each at an optimal field strength of 200 V/cm were administered to the animals using 10 mm Tweezertrodes (Genetronics, San Diego, CA, USA). The electrodes were placed on either side of the chest, which had been wetted with 70% ethanol. The animals recovered and survived with no apparent trauma until the experiments were terminated at the desired times, between 1 and 7 days post-treatment. Gene expression was detected by 1 day postelectroporation and peaked between 2 and 5 days. By 7 days, expression was back to baseline. By contrast, essentially no gene expression was detected in the absence of electric pulses. Using a beta-galactosidase-expressing plasmid, the distribution of gene expression appeared to be concentrated in the periphery of the lung, but was also present throughout the parenchyma. The primary cell types expressing gene product include alveolar type I and type II epithelial cells. No inflammation or lung injury was detected histologically or by cytokine measurements in lungs at either 1 or 24 h following electroporation treatment. These results provide evidence that electroporation is a safe and effective means for introducing naked DNA into the lung and form the basis for future studies on targeted pulmonary gene therapy.


Subject(s)
DNA/administration & dosage , Electroporation/methods , Genetic Therapy/methods , Lung Diseases/therapy , Lung/metabolism , Transfection/methods , Animals , Female , Gene Expression , Interleukin-6/analysis , Luciferases/genetics , Lung/immunology , Mice , Mice, Inbred BALB C
5.
J Biol Chem ; 276(45): 42485-91, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11551940

ABSTRACT

Peroxisome proliferators, which function as peroxisome proliferator-activated receptor alpha (PPARalpha) agonists, induce peroxisomal, microsomal, and mitochondrial fatty acid oxidation enzymes, in conjunction with peroxisome proliferation, in liver cells. Sustained activation of PPARalpha leads to the development of liver tumors in rats and mice. The assertion that synthetic PPARalpha ligands pose negligible carcinogenic risk to humans is attributable, in part, to the failure to observe peroxisome proliferation in human hepatocytes. To explore the mechanism(s) of species-specific differences in response to PPARalpha ligands, we determined the functional competency of human PPARalpha in vivo and compared its potency with that of mouse PPARalpha. Recombinant adenovirus that expresses human or mouse PPARalpha was produced and administered intravenously to PPARalpha-deficient mice. Human as well as mouse PPARalpha fully restored the development of peroxisome proliferator-induced immediate pleiotropic responses, including peroxisome proliferation and enhanced expression of genes involved in lipid metabolism as well as nonperoxisomal genes, such as CD36, Ly-6D, Rbp7, monoglyceride lipase, pyruvate dehydrogenase kinase-4, and C3f, that have been identified recently to be up-regulated in livers with peroxisome proliferation. These studies establish that human PPARalpha is functionally competent and is equally as dose-sensitive as mouse PPARalpha in inducing peroxisome proliferation within the context of mouse liver environment and that it can heterodimerize with mouse retinoid X receptor, and this human PPARalpha-mouse retinoid X receptor chimeric heterodimer transcriptionally activates mouse PPARalpha target genes in a manner qualitatively similar to that of mouse PPARalpha.


Subject(s)
Liver/metabolism , Peroxisomes/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Cell Division/drug effects , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Peroxisomes/drug effects , Pyrimidines/pharmacology , RNA, Messenger/analysis
6.
Proc Natl Acad Sci U S A ; 98(18): 10380-5, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517327

ABSTRACT

The nuclear receptor coactivators participate in the transcriptional activation of specific genes by nuclear receptors. In this study, we report the isolation of a nuclear receptor coactivator-interacting protein from a human liver cDNA library by using the coactivator peroxisome proliferator-activated receptor-interacting protein (PRIP) (ASC2/AIB3/RAP250/NRC/TRBP) as bait in a yeast two-hybrid screen. Human PRIP-interacting protein cDNA has an ORF of 2,556 nucleotides, encodes a protein with 852 amino acids, and contains a 9-aa VVDAFCGVG methyltransferase motif I and an invariant GXXGXXI segment found in K-homology motifs of many RNA-binding proteins. The gene encoding this protein, designated PRIP-interacting protein with methyltransferase domain (PIMT), is localized on chromosome 8q11 and spans more than 40 kb. PIMT mRNA is ubiquitously expressed, with a high level of expression in heart, skeletal muscle, kidney, liver, and placenta. Using the immunofluorescence localization method, we found that PIMT and PRIP proteins appear colocalized in the nucleus. PIMT strongly interacts with PRIP under in vitro and in vivo conditions, and the PIMT-binding site on PRIP is in the region encompassing amino acids 773-927. PIMT binds S-adenosyl-l-methionine, the methyl donor for methyltransfer reaction, and it also binds RNA, suggesting that it is a putative RNA methyltransferase. PIMT enhances the transcriptional activity of peroxisome proliferator-activated receptor gamma and retinoid-X-receptor alpha, which is further stimulated by coexpression of PRIP, implying that PIMT is a component of nuclear receptor signal transduction apparatus acting through PRIP. Definitive identification of the specific substrate of PIMT and the role of this RNA-binding protein in transcriptional regulation remain to be determined.


Subject(s)
Methyltransferases/genetics , Methyltransferases/metabolism , Protein D-Aspartate-L-Isoaspartate Methyltransferase , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Humans , Methyltransferases/chemistry , Mice , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , S-Adenosylmethionine/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Transcription Factors/metabolism , Transcription, Genetic , tRNA Methyltransferases/chemistry
7.
Gene Expr ; 9(4-5): 173-81, 2001.
Article in English | MEDLINE | ID: mdl-11444527

ABSTRACT

Spontaneous peroxisome proliferation-related pleiotropic responses occurring in the liver of mice lacking peroxisomal fatty acyl-CoA oxidase (AOX-/-) are attributed to sustained activation of peroxisome proliferator-activated receptor alpha (PPARalpha) by its putative natural ligands that require AOX for their metabolism. In this study, using a gene expression screen, we show that Ly-6 (lymphocyte antigen 6 complex, locus D; mouse ThB), which belongs to a distinctive family of low molecular weight phosphatidyl inositol anchored cell surface glycoproteins, is upregulated in mouse liver with peroxisome proliferation. Increases in Ly-6D mRNA levels are observed in AOX-/- mouse liver with spontaneous peroxisome proliferation and also in the liver of wild-type mice treated with synthetic peroxisome proliferators. Peroxisome proliferators failed to increase hepatic Ly-6D mRNA levels in mice lacking PPARalpha (PPARalpha-/-), suggesting a regulatory role for PPARalpha in the induction of Ly-6D. These observations suggest that changes in certain cell surface proteins also form part of the pleiotropic responses associated with peroxisome proliferation.


Subject(s)
Antigens, Ly/genetics , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Up-Regulation , Acyl-CoA Oxidase , Animals , Dose-Response Relationship, Drug , Gene Deletion , In Situ Hybridization , Liver/cytology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/genetics , Oxidoreductases/metabolism , Peroxisome Proliferators/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Up-Regulation/drug effects
8.
Exp Cell Res ; 268(1): 70-6, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11461119

ABSTRACT

Mice deficient in fatty acyl-CoA oxidase (AOX(-/-)), the first enzyme of the peroxisomal beta-oxidation system, develop specific morphological and molecular changes in the liver characterized by microvesicular fatty change, increased mitosis, spontaneous peroxisome proliferation, increased mRNA and protein levels of genes regulated by peroxisome proliferator-activated receptor alpha (PPARalpha), and hepatocellular carcinoma. Based on these findings it is proposed that substrates for AOX function as ligands for PPARalpha. In this study we examined the sequential changes in morphology and gene expression in the liver of wild-type and AOX(-/-) mice at Embryonic Day 17.5, and during postnatal development up to 2 months of age. In AOX(-/-) mice high levels of expression of PPARalpha-responsive genes in the liver commenced on the day of birth and persisted throughout the postnatal period. We found no indication of PPARalpha activation in the livers of AOX(-/-) mice at embryonic age E17.5. In AOX(-/-) mice microvesicular fatty change in liver cells was evident at 7 days. At 2 months of age livers showed extensive steatosis and the presence in the periportal areas of clusters of hepatocytes with abundant granular eosinophilic cytoplasm rich in peroxisomes. These results suggest that the biological ligands for PPARalpha vis a vis substrates for AOX either are not functional in fetal liver or do not cross the placental barrier during the fetal development and that postnatally they are likely derived from milk and diet.


Subject(s)
Isomerases , Liver/metabolism , Oxidoreductases/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Oxidase , Aging/metabolism , Animals , Animals, Newborn , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation, Developmental , Immunoblotting , In Situ Hybridization , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Liver/cytology , Liver/embryology , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Peroxisomal Bifunctional Enzyme , Peroxisomes/pathology , Peroxisomes/ultrastructure , RNA, Messenger/metabolism
9.
Gene Expr ; 9(6): 291-304, 2001.
Article in English | MEDLINE | ID: mdl-11764000

ABSTRACT

Peroxisome proliferators, which function as peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists, are a group of structurally diverse nongenotoxic hepatocarcinogens including the fibrate class of hypolipidemic drugs that induce peroxisome proliferation in liver parenchymal cells. Sustained activation of PPARalpha by these agents leads to the development of liver tumors in rats and mice. To understand the molecular mechanisms responsible for the pleiotropic effects of these agents, we have utilized the cDNA microarray to generate a molecular portrait of gene expression in the liver of mice treated for 2 weeks with Wy-14,643, a potent peroxisome proliferator. PPARalpha activation resulted in the stimulation of expression (fourfold or greater) of 36 genes and decreased the expression (fourfold or more decrease) of 671 genes. Enhanced expression of several genes involved in lipid and glucose metabolism and many other genes associated with peroxisome biogenesis, cell surface function, transcription, cell cycle, and apoptosis has been observed. These include: CYP2B9, CYP2B10, monoglyceride lipase, pyruvate dehydrogenase-kinase-4, cell death-inducing DNA-fragmentation factor-alpha, peroxisomal biogenesis factor 11beta, as well as several cell recognition surface proteins including annexin A2, CD24, CD39, lymphocyte antigen 6, and retinoic acid early transcript-gamma, among others. Northern blotting of total RNA extracted from the livers of PPARalpha-/- mice and from mice lacking both PPARalpha and peroxisomal fatty acyl-CoA oxidase (AOX), that were fed control and Wy-14,643-containing diets for 2 weeks, as well as time course of induction following a single dose of Wy-14,643, revealed that upregulation of genes identified by microarray procedure is dependent upon peroxisome proliferation vis-à-vis PPARalpha. However, cell death-inducing DNA-fragmentation factor-alpha mRNA, which is increased in the livers of wild-type mice treated with peroxisome proliferators, was not enhanced in AOX-/- mice with spontaneous peroxisome proliferation. These observations indicate that the activation of PPARalpha leads to increased and decreased expression of many genes not associated with peroxisomes, and that delayed onset of enhanced expression of some genes may be the result of metabolic events occurring secondary to PPARalpha activation and alterations in lipid metabolism.


Subject(s)
Liver/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , DNA, Complementary , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrimidines/pharmacology
10.
Biochem Biophys Res Commun ; 278(1): 250-7, 2000 Nov 11.
Article in English | MEDLINE | ID: mdl-11071880

ABSTRACT

The peroxisome proliferator-activated receptor alpha (PPAR alpha) is a nuclear receptor that transcriptionally regulates mitochondrial and peroxisomal fatty acid beta-oxidation enzymes in the liver. Ligands include synthetic peroxisome proliferators and some fatty acids. PPARalpha activation leads to predictable pleiotropic responses in liver including peroxisome proliferation, increased fatty acid oxidation, and hepatocellular carcinoma. In the current study, the response to PPAR alpha-activation was compared in the heart, kidney, and liver since the role of PPAR alpha in extrahepatic fatty acid-oxidizing organs has not been fully explored. Basal expression of mitochondrial beta-oxidation enzymes was comparable in the three tissues, but peroxisomal beta-oxidation enzymes were most abundant in the liver and less so in the kidney and especially in the heart. After PPAR alpha activation with ciprofibrate, both mitochondrial and peroxisomal beta-oxidation enzymes were induced, with the strongest response seen in the liver, a moderate response in the kidney, and no significant response in the heart. PPAR alpha mRNA analysis suggested that the differential response may be related to PPAR alpha expression.


Subject(s)
Clofibric Acid/analogs & derivatives , Fatty Acids/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Carnitine/metabolism , Cell Nucleus/metabolism , Clofibric Acid/pharmacology , Enzyme Activation , Fibric Acids , Gene Expression Regulation, Enzymologic , Kidney/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria, Liver/metabolism , Myocardium/metabolism , Oxygen/metabolism , Peroxisome Proliferators/pharmacology , Peroxisomes/metabolism , Rats , Ribonucleases/metabolism , Tissue Distribution , Transcription, Genetic
11.
J Biol Chem ; 275(37): 28918-28, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10844002

ABSTRACT

Fasting causes lipolysis in adipose tissue leading to the release of large quantities of free fatty acids into circulation that reach the liver where they are metabolized to generate ketone bodies to serve as fuels for other tissues. Since fatty acid-metabolizing enzymes in the liver are transcriptionally regulated by peroxisome proliferator-activated receptor alpha (PPARalpha), we investigated the role of PPARalpha in the induction of these enzymes in response to fasting and their relationship to the development of hepatic steatosis in mice deficient in PPARalpha (PPARalpha(-/-)), peroxisomal fatty acyl-CoA oxidase (AOX(-/-)), and in both PPARalpha and AOX (double knock-out (DKO)). Fasting for 48-72 h caused profound impairment of fatty acid oxidation in both PPARalpha(-/-) and DKO mice, and DKO mice revealed a greater degree of hepatic steatosis when compared with PPARalpha(-/-) mice. The absence of PPARalpha in both PPARalpha(-/-) and DKO mice impairs the induction of mitochondrial beta-oxidation in liver following fasting which contributes to hypoketonemia and hepatic steatosis. Pronounced steatosis in DKO mouse livers is due to the added deficiency of peroxisomal beta-oxidation system in these animals due to the absence of AOX. In mice deficient in AOX alone, the sustained hyperactivation of PPARalpha and up-regulation of mitochondrial beta-oxidation and microsomal omega-oxidation systems as well as the regenerative nature of a majority of hepatocytes containing numerous spontaneously proliferated peroxisomes, which appear refractory to store triglycerides, blunt the steatotic response to fasting. Starvation for 72 h caused a decrease in PPARalpha hepatic mRNA levels in wild type mice, with no perceptible compensatory increases in PPARgamma and PPARdelta mRNA levels. PPARgamma and PPARdelta hepatic mRNA levels were lower in fed PPARalpha(-/-) and DKO mice when compared with wild type mice, and fasting caused a slight increase only in PPARgamma levels and a decrease in PPARdelta levels. Fasting did not change the PPAR isoform levels in AOX(-/-) mouse liver. These observations point to the critical importance of PPARalpha in the transcriptional regulatory responses to fasting and in determining the severity of hepatic steatosis.


Subject(s)
Fasting , Fatty Acids/metabolism , Fatty Liver/etiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Lactic Acid/blood , Liver/metabolism , Liver Glycogen/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , RNA, Messenger/analysis , Triglycerides/metabolism
12.
J Biol Chem ; 275(18): 13510-6, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10788465

ABSTRACT

We previously isolated and identified steroid receptor coactivator-1 (SRC-1) and peroxisome proliferator-activated receptor (PPAR)-binding protein (PBP/PPARBP) as coactivators for PPAR, using the ligand-binding domain of PPARgamma as bait in a yeast two-hybrid screening. As part of our continuing effort to identify cofactors that influence the transcriptional activity of PPARs, we now report the isolation of a novel coactivator from mouse, designated PRIP (peroxisome proliferator-activated receptor interacting protein), a nuclear protein with 2068 amino acids and encoded by 13 exons. Northern analysis showed that PRIP mRNA is ubiquitously expressed in many tissues of adult mice. PRIP contains two LXXLL signature motifs. The amino-terminal LXXLL motif (amino acid position 892 to 896) of PRIP was found to be necessary for nuclear receptor interaction, but the second LXXLL motif (amino acid position 1496 to 1500) appeared unable to bind PPARgamma. Deletion of the last 12 amino acids from the carboxyl terminus of PPARgamma resulted in the abolition of the interaction between PRIP and PPARgamma. PRIP also binds to PPARalpha, RARalpha, RXRalpha, ER, and TRbeta1, and this binding is increased in the presence of specific ligands. PRIP acts as a strong coactivator for PPARgamma in the yeast and also potentiates the transcriptional activities of PPARgamma and RXRalpha in mammalian cells. A truncated form of PRIP (amino acids 786-1132) acts as a dominant-negative repressor, suggesting that PRIP is a genuine coactivator.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cloning, Molecular , Mediator Complex Subunit 1 , Mice , Molecular Sequence Data , Mutation , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism
13.
Mutat Res ; 448(2): 159-77, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10725470

ABSTRACT

Peroxisome proliferators are a structurally diverse group of non-genotoxic chemicals that induce predictable pleiotropic responses including the development of liver tumors in rats and mice. These chemicals interact variably with peroxisome proliferator-activated receptors (PPARs), which are members of the nuclear receptor superfamily. Evidence derived from mice with PPARalpha gene disruption indicates that of the three PPAR isoforms (alpha, beta/delta and gamma), the isoform PPARalpha is essential for the pleiotropic responses induced by peroxisome proliferators. Peroxisome proliferator-induced activation of PPARalpha leads to profound transcriptional activation of genes encoding for the classical peroxisomal beta-oxidation system and cytochrome P450 CYP 4A isoforms, CYP4A1 and CYP4A3, among others. Livers with peroxisome proliferation manifest substantial increases in the expression of H(2)O(2)-generating peroxisomal fatty acyl-CoA oxidase, the first enzyme of the classical peroxisomal fatty acid beta-oxidation system, and of microsomal cytochrome P450 4A1 and 4A3 genes. Disproportionate increases in H(2)O(2)-generating enzymes and H(2)O(2)-degrading enzyme catalase and reductions in glutathione peroxidase activity by peroxisome proliferators, lead to increased oxidative stress in liver cells. Sustained oxidative stress resulting from chronic increases in H(2)O(2)-generating enzymes manifests as massive accumulation of lipofuscin in hepatocytes, and increased levels of 8-hydroxydeoxyguanosine adducts in liver DNA; this supports the hypothesis that oxidative stress plays a critical role in the development of liver tumors induced by these non-genotoxic chemical carcinogens. Evidence also indicates that cells stably overexpressing H(2)O(2)-generating fatty acyl-CoA oxidase or urate oxidase, when exposed to appropriate substrate(s), reveal features of neoplastic conversion including growth in soft agar and formation of tumors in nude mice. Mice with disrupted fatty acyl-CoA oxidase gene (AOX(-/-) mice), which encodes the first enzyme of the PPARalpha regulated peroxisomal beta-oxidation system, exhibit profound spontaneous peroxisome proliferation, including development of liver tumors, indicative of sustained activation of PPARalpha by the unmetabolized substrates of acyl-CoA oxidase. With the exception of fatty acyl-CoA oxidase, all PPARalpha responsive genes including CYP4A1 and CYP4A3 are up-regulated in the livers of these AOX(-/-) mice. Thus, the substrates of acyl-CoA oxidase serve as endogenous ligands for this receptor leading to a receptor-enzyme cross-talk, because acyl-CoA oxidase gene is transcriptionally regulated by PPARalpha. Peroxisome proliferators induce only a transient increase in liver cell proliferation and this may serve as an additional contributory factor, rather than play a primary role in liver tumor development. Thus, sustained activation of PPARalpha by either synthetic or natural ligands leads to reproducible pleiotropic responses culminating in the development of liver tumors. This phenomenon of peroxisome proliferation provides fascinating challenges in exploring the molecular mechanisms of cell specific transcription, and in identifying the PPARalpha responsive target genes, as well as events involved in their regulation. Genetically altered animals and cell lines should enable investigations on the role of H(2)O(2)-producing enzymes in neoplastic conversion.


Subject(s)
Hydrogen Peroxide/metabolism , Liver Neoplasms/chemically induced , Peroxisome Proliferators/toxicity , Animals , Cell Transformation, Neoplastic , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Mice , Oxidoreductases/metabolism , Rats
14.
Proc Natl Acad Sci U S A ; 96(19): 10848-53, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10485914

ABSTRACT

Peroxisome proliferator-activated receptor binding protein (PBP), a nuclear receptor coactivator, interacts with estrogen receptor alpha (ERalpha) in the absence of estrogen. This interaction was enhanced in the presence of estrogen but was reduced in the presence of antiestrogen, tamoxifen. Transfection of PBP in CV-1 cells resulted in enhancement of estrogen-dependent transcription, indicating that PBP serves as a coactivator in ER signaling. To examine whether overexpression of PBP plays a role in breast cancer because of its coactivator function in ER signaling, we determined the levels of PBP expression in breast tumors. High levels of PBP expression were detected in approximately 50% of primary breast cancers and breast cancer cell lines by ribonuclease protection analysis, in situ hybridization, and immunoperoxidase staining. Fluorescence in situ hybridization of human chromosomes revealed that the PBP gene is located on chromosome 17q12, a region that is amplified in some breast cancers. We found PBP gene amplification in approximately 24% (6/25) of breast tumors and approximately 30% (2/6) of breast cancer cell lines, implying that PBP gene overexpression can occur independent of gene amplification. This gene comprises 17 exons that, together, span >37 kilobases. The 5'-flanking region of 2.5 kilobase pairs inserted into a luciferase reporter vector revealed that the promoter activity in CV-1 cells increased by deletion of nucleotides from -2,500 to -273. The -273 to +1 region, which exhibited high promoter activity, contains a typical CCAT box and multiple cis-elements such as C/EBPbeta, YY1, c-Ets-1, AP1, AP2, and NFkappaB binding sites. These observations, in particular PBP gene amplification, suggest that PBP, by its ability to function as ERalpha coactivator, might play a role in mammary epithelial differentiation and in breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Transcription Factors , Base Sequence , Chromosomes, Human, Pair 17 , Cloning, Molecular , Estrogen Receptor alpha , Exons , Humans , In Situ Hybridization, Fluorescence , Mediator Complex Subunit 1 , Molecular Sequence Data , Plasmids , Promoter Regions, Genetic , RNA, Messenger/analysis , Receptors, Estrogen/metabolism , Transfection , Tumor Cells, Cultured
15.
J Biol Chem ; 274(27): 19228-36, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383430

ABSTRACT

Fatty acid beta-oxidation occurs in both mitochondria and peroxisomes. Long chain fatty acids are also metabolized by the cytochrome P450 CYP4A omega-oxidation enzymes to toxic dicarboxylic acids (DCAs) that serve as substrates for peroxisomal beta-oxidation. Synthetic peroxisome proliferators interact with peroxisome proliferator activated receptor alpha (PPARalpha) to transcriptionally activate genes that participate in peroxisomal, microsomal, and mitochondrial fatty acid oxidation. Mice lacking PPARalpha (PPARalpha-/-) fail to respond to the inductive effects of peroxisome proliferators, whereas those lacking fatty acyl-CoA oxidase (AOX-/-), the first enzyme of the peroxisomal beta-oxidation system, exhibit extensive microvesicular steatohepatitis, leading to hepatocellular regeneration and massive peroxisome proliferation, implying sustained activation of PPARalpha by natural ligands. We now report that mice nullizygous for both PPARalpha and AOX (PPARalpha-/- AOX-/-) failed to exhibit spontaneous peroxisome proliferation and induction of PPARalpha-regulated genes by biological ligands unmetabolized in the absence of AOX. In AOX-/- mice, the hyperactivity of PPARalpha enhances the severity of steatosis by inducing CYP4A family proteins that generate DCAs and since they are not metabolized in the absence of peroxisomal beta-oxidation, they damage mitochondria leading to steatosis. Blunting of microvesicular steatosis, which is restricted to few liver cells in periportal regions in PPARalpha-/- AOX-/- mice, suggests a role for PPARalpha-induced genes, especially members of CYP4A family, in determining the severity of steatosis in livers with defective peroxisomal beta-oxidation. In age-matched PPARalpha-/- mice, a decrease in constitutive mitochondrial beta-oxidation with intact constitutive peroxisomal beta-oxidation system contributes to large droplet fatty change that is restricted to centrilobular hepatocytes. These data define a critical role for both PPARalpha and AOX in hepatic lipid metabolism and in the pathogenesis of specific fatty liver phenotype.


Subject(s)
Fatty Acids/metabolism , Fatty Liver/genetics , Microbodies/enzymology , Mitochondria/enzymology , Oxidoreductases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Acyl-CoA Oxidase , Animals , Blotting, Western , Fatty Liver/enzymology , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Phenotype
16.
Ann Plast Surg ; 42(6): 683-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10382809

ABSTRACT

Primary invasive Aspergillus Infection of the soft tissue is rare and typically affects immunocompromised patients in several distinct patterns of clinical presentation. In general, the role of surgery in the treatment of this disease is the removal of infected or necrotic tissue to prevent dissemination and mortality. However, the specific surgical recommendations have varied widely among reports due to the varied clinical circumstances in each series. The authors present the case of a patient with a primary invasive Aspergillus infection. They review the reported surgical experience with this disease, and discuss outcomes and surgical approaches in the context of several variations in clinical presentation. In all situations, antifungal therapy and prompt surgical intervention are critical in treating these initially localized but potentially lethal infections. The extent of intervention can range from minor debridement to amputation, and is based on the presence of persistent immunocompromise, the presence and extent of tissue necrosis, and the rate of progression during antifungal therapy.


Subject(s)
Aspergillosis/surgery , Foot Ulcer/microbiology , Foot Ulcer/surgery , Acquired Immunodeficiency Syndrome/complications , Adult , Aspergillosis/complications , Aspergillosis/immunology , Debridement , Humans , Immunocompromised Host , Male , Sarcoma, Kaposi/complications , Skin Neoplasms/complications , Skin Transplantation
17.
J Biol Chem ; 274(22): 15775-80, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336479

ABSTRACT

Peroxisomes contain a classical L-hydroxy-specific peroxisome proliferator-inducible beta-oxidation system and also a second noninducible D-hydroxy-specific beta-oxidation system. We previously generated mice lacking fatty acyl-CoA oxidase (AOX), the first enzyme of the L-hydroxy-specific classical beta-oxidation system; these AOX-/- mice exhibited sustained activation of peroxisome proliferator-activated receptor alpha (PPARalpha), resulting in profound spontaneous peroxisome proliferation in liver cells. These observations implied that AOX is responsible for the metabolic degradation of PPARalpha ligands. In this study, the function of enoyl-CoA hydratase/L-3-hydroxyacyl-CoA dehydrogenase (L-PBE), the second enzyme of this peroxisomal beta-oxidation system, was investigated by disrupting its gene. Mutant mice (L-PBE-/-) were viable and fertile and exhibited no detectable gross phenotypic defects. L-PBE-/- mice showed no hepatic steatosis and manifested no spontaneous peroxisome proliferation, unlike that encountered in livers of mice deficient in AOX. These results indicate that disruption of classical peroxisomal fatty acid beta-oxidation system distal to AOX step does not interfere with the inactivation of endogenous ligands of PPARalpha, further confirming that the AOX gene is indispensable for the physiological regulation of this receptor. The absence of appreciable changes in lipid metabolism also indicates that enoyl-CoAs, generated in the classical system in L-PBE-/- mice are diverted to D-hydroxy-specific system for metabolism by D-PBE. When challenged with a peroxisome proliferator, L-PBE-/- mice showed increases in the levels of hepatic mRNAs and proteins that are regulated by PPARalpha except for appreciable blunting of peroxisome proliferative response as compared with that observed in hepatocytes of wild type mice similarly treated. This blunting of peroxisome proliferative response is attributed to the absence of L-PBE protein in L-PBE-/- mouse liver, because all other proteins are induced essentially to the same extent in both wild type and L-PBE-/- mice.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , Enoyl-CoA Hydratase/genetics , Isomerases/genetics , Liver/enzymology , Microbodies/metabolism , Multienzyme Complexes/genetics , Acyl-CoA Oxidase , Animals , Catalase/metabolism , Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , Fatty Acids/metabolism , Fibric Acids , Mice , Mice, Knockout , Microbodies/genetics , Oxidoreductases/metabolism , Peroxisomal Bifunctional Enzyme , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 96(4): 1585-90, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-9990068

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors, and it is assumed that the biological effects of these receptors depend on interactions with recently identified coactivators, including steroid receptor coactivator-1 (SRC-1). We assessed the in vivo function of SRC-1 on the PPARalpha-regulated gene expression in liver by generating mice in which the SRC-1 gene was inactivated by gene targeting. The homozygous (SRC-1(-/-)) mice were viable and fertile and exhibited no detectable gross phenotypic defects. When challenged with a PPARalpha ligand, such as ciprofibrate or Wy-14,643, the SRC-1(-/-) mice displayed typical pleiotropic responses, including hepatomegaly, peroxisome proliferation in hepatocytes, and increased mRNA and protein levels of genes that are regulated by PPARalpha. These alterations were indistinguishable from those exhibited by SRC-1(+/+) wild-type mice fed either ciprofibrate- or Wy-14, 643-containing diets. These results indicate that SRC-1 is not essential for PPARalpha-mediated transcriptional activation in vivo and suggest redundancy in nuclear receptor coactivators.


Subject(s)
Gene Expression Regulation , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Cell Division , Clofibric Acid/analogs & derivatives , Clofibric Acid/pharmacology , DNA Primers , DNA-Binding Proteins/metabolism , Exons , Fibric Acids , Gene Expression Regulation/drug effects , Genotype , Histone Acetyltransferases , Liver/cytology , Mice , Mice, Inbred Strains , Mice, Knockout , Microbodies/drug effects , Microbodies/physiology , Nuclear Receptor Coactivator 1 , Pyrimidines/pharmacology , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/deficiency , Transcription Factors/genetics
19.
Gene Expr ; 8(4): 231-46, 1999.
Article in English | MEDLINE | ID: mdl-10794525

ABSTRACT

We have previously reported the cloning of rat deoxyuridine triphosphate nucleotidohydrolase (dUTPase) cDNA and demonstrated that the full-length protein as well as the N-terminal 62-amino acid peptide interacts with peroxisome proliferator-activated receptor alpha (PPARalpha). We now report the cloning of mouse dUTPase cDNA and show that it contains a 162-amino acid open reading frame, encoding a protein with a predicted Mr of 17,400 and differs from rat cDNA, which contains additional 43 amino acids at the N-terminal end. Unlike rat dUTPase, mouse dUTPase failed to bind PPARalpha. An evaluation of 205 amino acid containing rat dUTPase cDNA revealed that the N-terminal 43 extra amino acid segment contains an LXXLL signature motif, considered necessary and sufficient for the binding of several cofactors with nuclear receptors, and its absence in murine dUTPase possibly accounts for the differential binding of these enzymes to PPARalpha. In situ hybridization and immunohistochemical studies revealed that, in the adult mouse, dUTPase is expressed at high levels in proliferating cells of colonic mucosa, and of germinal epithelium in testis. At 9.5-day mouse embryonic development, dUTPase expression is predominantly in developing neural epithelium, and hepatic primordium, and in later developmental stages (11.5-, 13.5-, and 15.5-day embryo), the expression began to be localized to the liver, kidney, gut epithelium, thymus, granular layer of the cerebellum, and olfactory epithelium. We also show that the murine dUTPase gene comprises 6 exons and the 5'-flanking region of -1479 to -27, which exhibited high promoter activity, contains a typical TATA box and multiple cis-elements such as Sp-1, AP2, AP3, AP4, Ker1, RREB, and CREB binding sites. These observations suggest the existence of variants of dUTPase, some of which may influence nuclear receptor function during development and differentiation, in addition to catalyzing the hydrolysis of dUTP to dUMP.


Subject(s)
Pyrophosphatases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cloning, Molecular , Mice , Molecular Sequence Data , Organ Specificity , Pyrophosphatases/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Alignment , Sequence Analysis , Species Specificity
20.
Am J Pathol ; 153(2): 349-54, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9708794

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) regulate genes involved in lipid metabolism and adipocyte differentiation. Steroid receptor coactivator-1 (SRC-1) and PPAR-binding protein (PBP) interact with PPARgamma and act as coactivators to enhance ligand-dependent transcription. We report here that PPARgamma, SRC-1, and PBP are differentially expressed in the brown fat, transitional epithelium of the urinary bladder, colonic mucosa, and mammary epithelium of the adult mouse. PPARgamma and PBP are expressed in the transitional epithelium of urinary bladder and in brown adipose tissue, but not SRC-1. In the colonic mucosa, PPARgamma expression occurs throughout the villi, whereas the expression of both SRC-1 and PBP is confined mostly to the crypts. The expression of both SRC-1 and PBP is prominent in the breast epithelium of nonpregnant, pregnant, and lactating mice, whereas PPARgamma expression appeared prominent during lactation. During early embryonic development, PPARgamma, SRC-1, and PBP are differentially expressed, with only limited cell types displaying overlapping expression. PPARgamma and PBP expression overlapped in the brown fat and urogenital sinus at stage E15.5 of embryogenesis, whereas SRC-1 expression occurred mostly in neuroepithelium and cartilage between stages E9.5 and E13.5 of embryogenesis.


Subject(s)
Carrier Proteins/metabolism , Lactation/metabolism , Pregnancy, Animal/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Adipose Tissue, Brown/metabolism , Animals , Colon/metabolism , Embryo, Mammalian/metabolism , Female , Gene Expression , Gene Expression Regulation, Developmental , Histone Acetyltransferases , Immunohistochemistry , In Situ Hybridization , Intestinal Mucosa/metabolism , Mammary Glands, Animal/metabolism , Mediator Complex Subunit 1 , Mice , Mice, Inbred C57BL , Nuclear Receptor Coactivator 1 , Pregnancy , Time Factors , Tissue Distribution , Urinary Bladder/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...