Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Sci Med Sport ; 26(12): 707-710, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37951824

ABSTRACT

Interactions between statin therapy and physical exercise complicate effective cardiovascular prevention. Emerging evidence suggests that muscle strain related changes in the expression of the glucocorticoid-induced leucine zipper (GILZ) may be involved. Therefore, we measured GILZ mRNA expression levels in M. vastus lateralis samples of 32 healthy individuals before and after a standardized bout of strength or endurance exercise. Overall, we found a highly significant downregulation of GILZ after exercise training (p < 0.001). Within-subgroup changes were statistically significant only after strength training, supporting the role of muscle (as opposed to cardiocirculatory) strain. If confirmed, this may help fitting training recommendations and medication.


Subject(s)
Glucocorticoids , Transcription Factors , Humans , Glucocorticoids/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Leucine Zippers , Muscle, Skeletal/metabolism , Exercise
3.
Cell Death Dis ; 6: e1894, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26426686

ABSTRACT

Hepatocellular carcinoma (HCC) represents the third leading cause of cancer-related deaths and commonly develops in inflammatory environments. The IGF2 mRNA-binding protein IMP2-2/IGF2BP2-2/p62 was originally identified as an autoantigen in HCC. Aim of this study was to investigate a potential pathophysiological role of p62 in hepatocarcinogenesis. Human HCC tissue showed overexpression of IMP2, which strongly correlated with the fetal markers AFP and DLK1/Pref-1/FA-1 and was particularly elevated in tumors with stem-like features and hypervascularization. Molecular classification of IMP2-overexpressing tumors revealed an aggressive phenotype. Livers of mice overexpressing the IMP2 splice variant p62 highly expressed the stem cell marker DLK1 and secreted DLK1 into the blood. p62 was oncogenic: diethylnitrosamine (DEN)-treated p62 transgenic mice exhibited a higher tumor incidence and multiplicity than wild types. Tumors of transgenics showed a more aggressive and stem-like phenotype and displayed more oncogenic chromosomal aberrations determined with aCGH analysis. DEN-treated p62 transgenic mice exhibited distinct signs of inflammation, such as inflammatory cytokine expression and oxidative stress markers, that is, thiobarbituric acid-reactive substance (TBARS) levels. Reactive oxygen species (ROS) production was elevated in HepG2 cells, which either overexpressed p62 or were treated with DLK1. p62 induced this ROS production by a DLK1-dependent induction and activation of the small Rho-GTPase RAC1, activating NADPH oxidase and being overexpressed in human HCC. Our data indicate that p62/IMP2 promotes hepatocarcinogenesis by an amplification of inflammation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms, Experimental/genetics , Lung Neoplasms/genetics , RNA-Binding Proteins/genetics , Animals , Calcium-Binding Proteins , Carcinoma, Hepatocellular/secondary , Genomic Instability , Hep G2 Cells , Humans , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Liver Neoplasms, Experimental/pathology , Lung Neoplasms/secondary , Mice, Transgenic , Neoplastic Stem Cells/physiology , Neuropeptides/metabolism , Oxidative Stress , Phenotype , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/metabolism
4.
Eur Surg Res ; 39(6): 325-31, 2007.
Article in English | MEDLINE | ID: mdl-17622730

ABSTRACT

BACKGROUND/AIM: The aim of the study was to characterize the hepatic injury (HI) of the nonischemic liver lobe after selective portal triad clamping and investigate the influence of pharmacological pretreatment with alpha-lipoic acid (LA). METHODS: Brown-Norway rats received 500 micromol LA injected via the inferior vena cava 15 min prior to the induction of 90 min of selective ischemia. Another group of rats received vehicle prior to ischemia. Both groups were compared with sham-operated animals. RESULTS: Lipid peroxidation (LPO) was increased in the ischemic as well as in the nonischemic liver tissue (NIL) in the untreated group. Levels of adenosine triphosphate and reduced glutathione content of the nonischemic liver lobe were decreased in the untreated group 1 h after reperfusion. Activity of caspases 3 and 8 was not detectable, whereas expression of the Bax protein was demonstrated in the NIL. We observed areas of necrotic hepatocytes and large gaps of sinusoids in the NIL of the untreated rats. LA attenuated LPO as well as Bax expression in the NIL. Moreover adenosine triphosphate and glutathione content of the NIL was increased 1 h after reperfusion by LA. LA pretreatment reduced release of alpha-glutathione-s-transferase in plasma. Histology of the nonischemic liver lobe did not markedly differ from sham-operated animals after LA pretreatment. CONCLUSION: HI of the NIL seems to be mediated by LPO and proapoptotic proteins such as Bax. Besides its described potential to reduce ischemia/reperfusion injury of the ischemic lobe, LA attenuates HI of the nonischemic tissue after selective portal triad clamping.


Subject(s)
Antioxidants/pharmacology , Ischemia/complications , Liver Diseases/prevention & control , Liver/drug effects , Thioctic Acid/pharmacology , Animals , Antioxidants/therapeutic use , Constriction , Disease Models, Animal , Lipid Peroxidation/drug effects , Liver/blood supply , Liver Diseases/etiology , Male , Rats , Reperfusion Injury/etiology , Reperfusion Injury/prevention & control , Thioctic Acid/therapeutic use
5.
Eur Surg Res ; 38(6): 503-12, 2006.
Article in English | MEDLINE | ID: mdl-17028433

ABSTRACT

BACKGROUND/AIM: Alpha-lipoic (LA) acid pretreatment has previously been described to reduce ischemia/reperfusion injury (IRI) after warm liver ischemia, whereas glycine pretreatment has been shown to be protective mostly in models of cold hepatic ischemia. The aim of this study was to determine whether glycine decreases IRI after warm hepatic ischemia. Furthermore we investigated whether doses of LA other than those used previously are also protective against IRI after warm hepatic ischemia. METHODS: Selective liver ischemia was maintained over a period of 90 min. In long-term as well as short-term experiments we studied IRI in several groups comparing animal survival as the pivotal endpoint. RESULTS: Animal survival was improved by glycine and 5,000 micromol LA, whereas all animals died within 3 days after pretreatment with 50 micromol LA. In the glycine group we observed a tendency towards decreased apoptosis-related cell death measured by the activity of caspase-3 in liver tissue and the percentage of TUNEL-positive hepatocytes in comparison to the untreated group. Serum alpha-glutathione S-transferase, lipid peroxidation, and caspase-3 activity as well as the percentage of TUNEL-positive hepatocytes and the percentage of liver necrosis were only significantly decreased by 5,000 micromol LA pretreatment. Liver tissue levels of tumor necrosis factor (TNF)alpha were reduced only in the glycine group whereas TNFalpha was increased in the untreated as well as the LA group. Levels of TNFalpha mRNA were upregulated in both the glycine- and LA-pretreated groups. CONCLUSION: Our data show that increased animal survival by glycine was accompanied by a reduced TNFalpha content in liver tissue. Protection by glycine is likely to result from a reduction in adverse TNFalpha effects. Administration of high-dose LA on the other hand led to a significant reduction in necrosis- and apoptosis-related cell death in IRI of the liver without a reduction in liver TNFalpha.


Subject(s)
Cytoprotection , Glycine/therapeutic use , Liver/blood supply , Reperfusion Injury/prevention & control , Thioctic Acid/therapeutic use , Warm Ischemia , Animals , Apoptosis/drug effects , Glutathione Transferase/metabolism , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Lipoxygenase/analysis , Liver/enzymology , Male , RNA, Messenger/analysis , Rats , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics
6.
Oncogene ; 20(46): 6679-88, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11709702

ABSTRACT

The adenovirus E1A12S gene product (WT12S) immortalizes epithelial cells and they retain their differentiated characteristics, but certain mutants cannot do the latter. Characterization of mutant immortalized epithelial cells indicated that they had undergone epithelial mesenchymal transition (EMT). Coexpression of V12ras with WT12S leads to benign tumors, but to malignant tumors with 12S mutants. Since EMT is critical for tumor progression, identification of the molecular mechanisms involved should elucidate novel therapeutic targets. To this end, representational difference analysis (RDA) was used to identify cDNAs upregulated in the mutant cell line. Thirty-five differentially expressed mRNAs were identified and classified into several functional categories, including nine novel cDNAs. Among the 26 known cDNAs, extracellular matrix and related proteins made up the largest group of differentially expressed genes, followed by growth factors and receptors and transcription factors. There was also an ion transporter, a cytoskeletal protein, glycosylation and amidinotransferase enzymes and proteins with unknown functions. Some of the known genes have previously been associated with EMT and/or tumor progression and thus served to validate the system to obtain the desired target genes, while other cDNAs are newly linked with dedifferentiation/malignancy. Array analyses indicated that the cDNAs were specifically upregulated in invasive or metastatic tumors, especially of breast, uterus and lung, suggesting their involvement in the progression of these tumors.


Subject(s)
Epithelium/embryology , Mesoderm/metabolism , Mesoderm/physiology , Adenoviridae/genetics , Cell Adhesion , Cell Line , Cytoskeleton/metabolism , DNA/metabolism , DNA, Complementary/metabolism , Disease Progression , Epithelial Cells/metabolism , Epithelium/metabolism , Epithelium/physiology , Extracellular Matrix/metabolism , Glycosylation , Humans , Models, Biological , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Protein Binding , Proteoglycans/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Mol Pharmacol ; 60(3): 421-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502871

ABSTRACT

BACKGROUND: The inducible nitric-oxide synthase (iNOS) synthesizes NO from L-arginine. Availability of L-arginine is maintained by a lipopolysaccharide (LPS)-induced induction of the CAT-2B amino acids transporter. Recently, we could show that the cardiovascular hormone atrial natriuretic peptide (ANP) inhibits the induction of iNOS in LPS-stimulated macrophages via its guanylate cyclase-coupled A-receptor. PURPOSE: To investigate whether ANP exerts an effect on LPS-induced L-arginine uptake. METHODS: Murine bone marrow derived macrophages were activated with LPS (1 microg/ml, 20 h) in the presence or absence of ANP or C-type natriuretic peptide (CNP). L-Arginine transport was determined by measuring the uptake of L-[(3)H]arginine. L-[(3)H]Arginine influx was also determined in LPS-activated cells in the presence of N(G)-monomethyl-L-arginine (L-NMMA), competitor amino acids, or ANP. Nitrite accumulation was determined in supernatants of LPS-activated cells cultured in the presence or absence of L-ornithine. RESULTS: ANP dose dependently (10(-8)-10(-6)M) inhibited LPS-induced L-[(3)H]arginine uptake when added simultaneously with LPS, whereas it showed no effect when added simultaneously with L-[(3)H]arginine. The effect was abrogated by the A-receptor antagonist HS-142-1 (10 microg/ml). CNP (10(-6) M) did not influence L-arginine transport. Competitor amino acids (10(-2) M) inhibited L-[(3)H]arginine uptake. An excess of unlabeled L-arginine (10(-2) M) as well as its analog L-NMMA (10(-3) M) also reduced L-[(3)H]arginine influx. L-Arginine uptake was critical for production of NO because L-ornithine significantly decreased LPS-induced nitrite accumulation. CONCLUSION: This work demonstrates that ANP inhibits LPS-induced L-arginine uptake via its guanylate cyclase-coupled A-receptor. Besides its influence on the induction of iNOS, this effect may represent an important and unique mechanism by which ANP regulates NO production in macrophages.


Subject(s)
Arginine/metabolism , Atrial Natriuretic Factor/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Binding, Competitive , Biological Transport/drug effects , Cells, Cultured , Drug Interactions , Guanylate Cyclase/metabolism , Hormones/pharmacology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase Type II , Receptors, Atrial Natriuretic Factor/metabolism , Substrate Specificity , Tritium
8.
Hepatology ; 34(3): 535-47, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526540

ABSTRACT

Liver resident NK1.1+ T cells are supposed to play a pivotal role in the onset of inflammatory liver injury in experimental mouse models such as concanavalin A (Con A)-induced hepatitis. These cells, expressing the adhesion receptor, CD44, are largely depleted from the liver by a single intravenous injection of low-molecular-weight fragments of hyaluronic acid (LMW-HA). Here, we report that LMW-HA pretreatment protected mice from liver injury in several models of T-cell- and macrophage-dependent, tumor necrosis factor alpha (TNF-alpha)-mediated inflammatory liver injury, i.e., from liver injury induced by either Con A or Pseudomonas exotoxin A (PEA) or PEA/lipopolysaccharide (LPS). Interestingly, apart from inhibition of cellular adhesion, pretreatment of mice with LMW-HA was also capable of preventing hepatocellular apoptosis and activation of caspase-3 induced by direct administration of recombinant murine (rmu) TNF-alpha to D-galactosamine (GalN)-sensitized mice. LMW-HA-induced hepatoprotection could be neutralized by pretreatment with the nuclear factor-kappaB (NF-kappaB) inhibitor, pyrrolidine dithiocarbamate (PDTC), demonstrating the involvement of NF-kappaB in the observed protective mechanism. Indeed, injection of LMW-HA rapidly induced the production of TNF-alpha by Kupffer cells and the translocation of NF-kappaB into hepatocellular nuclei. Both LMW-HA-induced TNF-alpha production and NF-kappaB translocation were blocked by pretreatment with PDTC. Our findings provide evidence for an unknown mechanism of LMW-HA-dependent protection from inflammatory liver disease, i.e., induction of TNF-alpha- and NF-kappaB-dependent cytoprotective proteins within the target parenchymal liver cells.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Chemical and Drug Induced Liver Injury/prevention & control , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha , Virulence Factors , Animals , CD4 Lymphocyte Count , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/pharmacology , Cytokines/blood , Exotoxins/pharmacology , Hyaluronan Receptors/analysis , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/pathology , Liver Failure/etiology , Liver Failure/prevention & control , Lymphocyte Count , Macrophages/physiology , Male , Mice , Mice, Inbred BALB C , Molecular Weight , NF-kappa B/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Pseudomonas aeruginosa Exotoxin A
9.
Immunol Cell Biol ; 79(1): 11-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11168617

ABSTRACT

Atrial natriuretic peptide (ANP) attenuates LPS-induced inducible nitric oxide synthase (iNOS) expression in murine macrophages by destabilizing iNOS mRNA. Because elevated intracellular free Ca2+ levels [Ca2+]i reduce iNOS mRNA stability, the aim of the present study was to determine whether inhibition of iNOS by ANP is due to alterations in intracellular calcium. As determined by fluorescence photometry, ANP (10(-7) and 10(-6) mol/L) was shown to elevate intracellular calcium levels in bone marrow-derived macrophages. This effect seemed to be mediated via the guanylate cyclase-coupled A receptor, because dibutyryl-cGMP mimicked and the A-receptor antagonist HS-142-1 partially abrogated the effect of ANP. Because the Ca2+ increase was also observed in Ca2+-free buffer, it is suggested that the liberation of intracellular calcium pools contributes to the elevation of [Ca2+]i by ANP. The B-receptor ligand C-type natriuretic peptide (CNP), which does not alter iNOS expression, had no effect on [Ca2+]i. The Ca2+-ionophore 4-Br-A23187 and thapsigargin, a compound known to liberate Ca2+ from intracellular stores, were further demonstrated to reduce LPS-induced NO production in macrophages (Griess assay), confirming a functional link for elevated [Ca2+]i and iNOS inhibition. These effects were abrogated by coincubation with extra- as well as intracellular Ca2+ chelators (EGTA, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA)). The inhibitory effect of ANP on NO production was also abrogated by Ca2+ chelation. These findings support a causal relationship between reduced iNOS induction and elevation of [Ca2+]i. Taken together, the data indicate that intracellular Ca2+ elevation by ANP is involved in the inhibition of LPS-induced nitric oxide production in macrophages.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Calcimycin/analogs & derivatives , Calcium/metabolism , Macrophages/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/biosynthesis , Aniline Compounds/metabolism , Animals , Atrial Natriuretic Factor/antagonists & inhibitors , Calcimycin/pharmacology , Cells, Cultured , Dibutyryl Cyclic GMP/pharmacology , Fluorescent Dyes/metabolism , Guanylate Cyclase/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Polysaccharides/pharmacology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Thapsigargin/pharmacology , Xanthenes/metabolism
10.
Ann Rheum Dis ; 60 Suppl 3: iii68-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11890659

ABSTRACT

The atrial natriuretic peptide (ANP), a member of the natriuretic peptide family, is a cardiovascular hormone which possesses well defined natriuretic, diuretic, and vasodilating properties. Most of the biological effects of ANP aremediated through its guanylyl cyclase coupled A receptor. Because ANP and its receptors have been shown to be expressed and differentially regulated in the immune system, it has been suggested that ANP has an immunomodulatory potency. Much investigation of the effects of ANP on the activation of macrophages has been carried out. ANP was shown to inhibit the lipopolysaccharide (LPS)-induced expression of inducible nitric oxide synthase (iNOS) in macrophages in an autocrine fashion. ANP in this context was shown to reduce significantly the activation of NF-kappaB and to destabilise iNOS mRNA. ANP, furthermore, can significantly reduce the LPS-induced secretion of tumour necrosis factor alpha (TNFalpha) in macrophages. The relevance of these findings on a regulatory role for ANP on TNFalpha in humans was shown by the fact that ANP significantly reduces the release of TNFalpha in whole human blood. It was furthermore shown to attenuate the release of interleukin 1beta (IL1beta). Interestingly, ANP did not affect the secretion of the anti-inflammatory cytokines IL10 and IL1 receptor antagonist (IL1ra). In summary, ANP was shown to reduce the secretion of inflammatory mediators in macrophages. Therefore, this cardiovascular hormone may possess anti-inflammatory potential.


Subject(s)
Atrial Natriuretic Factor/physiology , Autocrine Communication/physiology , Inflammation Mediators/metabolism , Macrophages/immunology , Rheumatic Diseases/immunology , Humans , Macrophage Activation/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Tumor Necrosis Factor-alpha/biosynthesis
11.
Crit Rev Immunol ; 21(6): 473-85, 2001.
Article in English | MEDLINE | ID: mdl-12058861

ABSTRACT

The atrial natriuretic peptide (ANP) was described as a peptide hormone synthesized and secreted by heart atria. It plays an important role in the regulation of volume homeostasis; however, the functions of ANP are not restricted to cardiovascular effects. The biological profile of ANP is much broader than originally thought. This article focuses on the immunomodulatory and anti-inflammatory functions of ANP addressing; for example, the influence of ANP on macrophage functions. Another important aspect of ANP reviewed here is its cytoprotective potential. The beneficial effect of ANP in preventing cell damage caused by ischemia and reperfusion warrants special attention. The therapeutic potential of ANP in organ preservation could be important for transplantation medicine.


Subject(s)
Adjuvants, Immunologic , Atrial Natriuretic Factor/immunology , Amino Acid Sequence , Animals , Atrial Natriuretic Factor/genetics , Gene Expression Regulation , Humans , Immune System , Molecular Sequence Data
12.
J Hepatol ; 33(2): 236-46, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10952241

ABSTRACT

BACKGROUND/AIMS: The cardiovascular hormone Atrial Natriuretic Peptide (ANP) attenuates activation of the pro-inflammatory transcription factor NF-kappaB in macrophages. ANP was also shown to protect from ischemia-reperfusion injury of the rat liver. This study aimed to investigate the effects of this immunomodulatory hormone and its second messenger cGMP on the activation of the two redox-sensitive transcription factors AP-1 and NF-kappaB and the expression of corresponding pro-inflammatory target genes during ischemia and reperfusion of the liver. The identification of the mechanisms underlying the protection by ANP should reveal new aspects concerning the pathomechanisms of ischemia/reperfusion injury. METHODS: Rat livers were perfused with and without ANP or 8-Br-cGMP preceding 24 h of cold storage in University of Wisconsin solution. During reperfusion NF-kappaB and AP-1 DNA binding activities were determined in freeze-clamped liver samples by electrophoretic mobility shift assay. Protein levels of p50, p65, and of IkappaB were determined by Western blot. mRNA coding for inducible nitric oxide synthase, cyclooxygenase-2, and TNF-alpha was determined by RT-PCR and Northern blot. RESULTS: After 45 min of reperfusion DNA binding activities of NF-kappaB were increased, whereas in ANP pre-treated livers this effect was markedly reduced. AP-1, another important redox-sensitive transcription factor, was activated and in the course of reperfusion the subunit composition of AP-1 changed as assessed by supershift assays. ANP markedly reduced binding activities of both forms of AP-1. 8-Br-cGMP mimicked the effects of ANP on NF-kappaB and AP-1. Neither inducible nitric oxide synthase nor cyclooxygenase-2 mRNA could be detected. In contrast, a profound expression of transcripts coding for TNF-alpha was detected in the course of reperfusion and ANP markedly reduced TNF-alpha mRNA expression. CONCLUSION: ANP seems to mediate its protective effect during ischemia and reperfusion by reducing the activation of NF-kappaB and AP-1 via cGMP. The reduced binding activity of these redox-sensitive transcription factors was accompanied by a diminished mRNA expression of TNF-alpha, a cytokine known to be involved in cellular damage in ischemia reperfusion injury.


Subject(s)
Atrial Natriuretic Factor/administration & dosage , Ischemia/metabolism , Liver/blood supply , Liver/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Down-Regulation/drug effects , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reperfusion , Reperfusion Injury/metabolism
13.
J Immunol ; 165(1): 175-81, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861050

ABSTRACT

The atrial natriuretic peptide (ANP) is suggested to regulate inflammatory response by alteration of macrophage functions. The aim of this study was to investigate whether ANP influences production of TNF-alpha. TNF-alpha production in murine bone marrow-derived macrophages was induced by LPS, and TNF-alpha secretion (+/-ANP) was determined by L929 bioassay. ANP dose dependently (10-8-10-6 M) inhibited TNF-alpha release by up to 95%. The effect was mediated via the guanylate cyclase-coupled A receptor, as was shown by employing dibutyryl-cGMP, the cGMP-inhibitory compound Ly-83583, and the A receptor antagonist HS-142-1. A specific ligand of the natriuretic peptide "clearance" receptor inhibited TNF-alpha production only at 10-7 and 10-8 M, but not at 10-6 M. The B receptor ligand C-type natriuretic peptide showed no TNF-alpha-inhibitory effect. To investigate the underlying mechanism of ANP-mediated TNF-alpha inhibition, Northern blot was performed. ANP-treated macrophages displayed decreased TNF-alpha-mRNA levels. Besides the known inhibition of NF-kappaB activation, in this study we demonstrated that ANP also attenuates the activation of the proinflammatory transcription factor AP-1 (gel shift assay). ANP did not alter subunit composition of AP-1 complexes, as was shown by supershift assays applying anti-c-jun and anti-c-fos Abs. To get information on the ANP effect for human inflammatory processes, we investigated cytokine production in human LPS-activated blood. ANP significantly attenuated production of TNF-alpha and IL-1beta without affecting production of IL-10 and IL-1ra. In summary, ANP was shown to attenuate TNF-alpha production of LPS-activated macrophages via cGMP. The inhibition is suggested to involve transcriptional processes that are the result of reduced activation of responsible transcription factors.


Subject(s)
Atrial Natriuretic Factor/antagonists & inhibitors , Atrial Natriuretic Factor/physiology , Cyclic GMP/physiology , Immunosuppressive Agents , Macrophages/immunology , Macrophages/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Atrial Natriuretic Factor/pharmacology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/blood , DNA-Binding Proteins/metabolism , Humans , Immunosuppressive Agents/pharmacology , Macromolecular Substances , Mice , Natriuretic Peptide, C-Type/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/physiology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/genetics
14.
J Pharmacol Exp Ther ; 287(3): 1098-104, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9864298

ABSTRACT

The seleno-organic drug ebselen (2-phenyl-1, 2-benzoisoselenazol-3(2H)-one) has glutathione peroxidase-like activity, and inhibits lipoxygenases, oxidative burst of leukocytes, nitric oxide synthases, protein kinases and leukocyte migration. This study elaborates in vivo in mice hitherto unknown immunopharmacological properties of ebselen. The compound was comparatively investigated in two different T cell-dependent hepatic hyperinflammation models and in two alternative models of receptor-activated liver apoptosis. Mice orally pretreated with ebselen were dose-dependently protected from concanavalin A (ConA)-induced liver injury. In livers from ebselen-pretreated mice exposed to ConA, the nuclear antiapoptotic transcription factor NFkappaB was upregulated. The release of the proinflammatory cytokine tumor necrosis factor-alpha (TNF) was downregulated, while the ciculating amount of the anti-inflammatory cytokine interleukin-10 (IL-10) was increased. Ebselen protected also from liver injury induced by the superantigen staphylococcal enterotoxin B in galactosamine (GalN)-sensitized mice. Furthermore, ebselen protected the liver and enhanced circulating IL-10 in GalN-sensitized mice treated with recombinant TNF, i.e., the common distal mediator of ConA and SEB-induced hepatotoxicity. The activation of apoptosis-executing proteases, i.e., caspases, was blocked in livers of ebselen-treated mice following TNF receptor, but not following CD95 receptor activation. We propose a novel mechanism for the immunomodulatory properties of the drug and suggest that it might be useful in the therapy of T cell-mediated inflammatory disorders.


Subject(s)
Azoles/pharmacology , Immunosuppressive Agents/pharmacology , Liver/drug effects , Organoselenium Compounds/pharmacology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Caspases/metabolism , Concanavalin A , Enterotoxins , Enzyme Activation , Isoindoles , Liver/metabolism , Liver/pathology , Liver Failure/prevention & control , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
15.
Hepatology ; 28(5): 1309-17, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9794916

ABSTRACT

The aim of our studies was to investigate hormonal prevention of hepatic preservation damage by the atrial natriuretic peptide (ANP) and the mechanisms involved. Isolated perfusion of rat livers was performed in a nonrecirculating fashion. Twenty minutes of preischemic perfusion was performed with or without different concentrations of ANP, followed by 24-hour storage in cold University of Wisconsin (UW) solution. Two hundred nanomoles of ANP prevented hepatocellular damage during a 2-hour reperfusion period as indicated by a marked attenuation of the sinusoidal efflux of lactate dehydrogenase (LDH) and purine nucleoside phosphorylase (PNP), and by reduced Trypan blue uptake. Furthermore, postischemic bile flow as an indicator of liver function was significantly improved by about 60% with 200 nmol/L ANP. No protection was conveyed by 20 nmol/L ANP nor by pretreatment with 200 nmol/L ANP for only 10 minutes. The effects of ANP seemed to be mediated by the guanylate cyclase-coupled A (GC-A) receptor and cyclic guanosine monophosphate (cGMP): whereas expression of both GC-A and GC-B receptors as well as of the GC-C receptor was found, cGMP did protect from ischemia-reperfusion damage, but selective ligands of the B and C receptor did not. To begin to determine the mechanisms of ANP-mediated protection, different parameters were investigated: ANP had no effect on portal pressure as an indicator of hepatic circulation, nor on intracellular energy depletion determined by adenosine nucleotide concentration. However, the marked augmentation of nuclear factor kappaB (NF-kappaB) binding activity during reperfusion was prevented in ANP-pretreated livers. In conclusion, pretreatment with ANP protects the rat liver from cold ischemia-reperfusion damage. This effect is mediated via the GC-A receptor and cGMP, and may be linked to an influence of ANP on NF-kappaB activation. Thus, ANP signaling via the GC-A receptor should be considered as a new pharmacological target to prevent preservation injury of the liver.


Subject(s)
Cold Temperature , Guanylate Cyclase/metabolism , Ischemia , Liver/blood supply , Receptors, Atrial Natriuretic Factor/physiology , Reperfusion Injury/prevention & control , Animals , Atrial Natriuretic Factor/therapeutic use , Cyclic GMP/analogs & derivatives , Cyclic GMP/physiology , Cyclic GMP/therapeutic use , L-Lactate Dehydrogenase/metabolism , Male , NF-kappa B/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Rats , Rats, Sprague-Dawley
16.
Atherosclerosis ; 139(2): 333-9, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9712340

ABSTRACT

Inducible nitric oxide synthase (iNOS) has recently been shown to be present in human atherosclerotic lesions and to promote the formation of deleterious peroxynitrite. Allicin and ajoene are discussed as active compounds with regard to the beneficial effects of garlic in atherosclerosis. The aim of this study was to investigate the effect of allicin and ajoene on the iNOS system in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Ajoene (IC50 2.5-5 microM) and allicin (IC50 15-20 microM) dose dependently reduced nitrite accumulation, a parameter for NO synthesis, in supernatants of LPS-stimulated (1 microg/ml, 20 h) macrophages. Accordingly, reduced iNOS enzyme activities were measured by conversion of L-[3H]arginine to L-[3H]citrulline in homogenates of LPS-activated cells treated with ajoene or allicin. None of these compounds, however, showed a direct effect on the catalytic-activity of iNOS. Consequently, iNOS protein and mRNA expression in ajoene (10 microM) or allicin (50 microM) treated cells were evaluated by Western blot and Northern blot analysis, respectively. Markedly reduced iNOS protein as well as mRNA levels were demonstrated. These observations indicate that allicin and ajoene inhibit the expression of iNOS in activated macrophages. The possible link of this effect to the beneficial features attributed to garlic is discussed.


Subject(s)
Disulfides/pharmacology , Garlic/chemistry , Macrophages/metabolism , Nitric Oxide Synthase/metabolism , Plant Extracts/pharmacology , Plants, Medicinal , Sulfinic Acids/pharmacology , Cell Line , Dose-Response Relationship, Drug , Homeostasis/physiology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Sulfoxides
17.
J Biol Chem ; 273(22): 13444-51, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9593677

ABSTRACT

Atrial natriuretic peptide (ANP), a cardiovascular hormone, has been shown to inhibit synthesis of nitric oxide in lipopolysaccharide (LPS)-activated mouse bone marrow-derived macrophages via activation of its guanylate cyclase-coupled receptor. The goal of the present study was to elucidate the potential sites of inducible nitric-oxide synthase (iNOS) regulation affected by ANP and revealed the following. 1) ANP and dibutyryl-cGMP did not inhibit catalytic iNOS activity measured by the conversion rate of L-[3H]arginine to L-[3H]citrulline in homogenates of LPS-treated cells. 2) Pretreatment of cells with ANP dose-dependently reduced the LPS-induced L-[3H]citrulline production that has been shown to be due to reduced iNOS protein levels detected by Western blot. 3) ANP does not alter the ratio of catalytically active iNOS dimer versus inactive iNOS monomer considered to be a major post-translational regulatory mechanism for the enzyme. 4) Macrophages exposed to ANP display decreased LPS-induced iNOS mRNA levels. 5) Importantly, two basic mechanisms seem to be responsible for this observation, i.e. ANP specifically induced acceleration of iNOS mRNA decay and ANP reduced binding activity of NF-kappaB, the transcription factor predominantly responsible for LPS-induced iNOS expression in murine macrophages. Moreover, 6) ANP acts via an autocrine mechanism since recently ANP was shown to be secreted by LPS-activated macrophages, and we demonstrated here that LPS-induced NO synthesis was increased after blocking the binding of endogenous ANP by a receptor antagonist. These observations suggest ANP as a new autocrine macrophage factor regulating NO synthesis both transcriptionally and post-transcriptionally. ANP may help to balance NO production of activated macrophages and thus may allow successful immune response without adverse effects on host cells.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Bone Marrow Cells/drug effects , Macrophages/drug effects , Nitric Oxide Synthase/metabolism , Animals , Bone Marrow Cells/enzymology , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Guanylate Cyclase/metabolism , Macrophages/enzymology , Mice , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/metabolism
18.
Endocrinology ; 138(10): 4282-90, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9322941

ABSTRACT

Atrial natriuretic peptide (ANP) has previously been suggested to inhibit the production of NO in LPS-activated primary macrophages. The aim of the present study was 1) to examine whether ANP elicits this effect also on macrophage cell lines (RAW 264.7, J774), 2) to elucidate whether ANP is the only natriuretic peptide (NP) inhibiting NO synthesis, 3) to look for the expression of natriuretic peptide receptors (NPR) on macrophages, 4) to consequently determine the type of receptor mediating the ANP effect and 5) to obtain first information on the underlying mechanism. Whereas ANP dose dependently (10(-6)-10(-8) M) inhibited NO synthesis (measured as nitrite accumulation, 20h) in all four types of macrophages (bone marrow derived and peritoneal macrophages; RAW 264.7 and J 774), urodilatin and atriopeptin I displayed only a weak effect restricted to the highest concentration (10(-6) M) employed. Importantly, C-type natriuretic peptide (CNP) showed no NO-inhibitory effect. The lack of effect of CNP was shown not to be due to its lower stability or its missing receptor. Macrophages were shown to express all three natriuretic peptide receptors (NPR-A, NPR-B, NPR-C) using RT-PCR technique. Furthermore, two types of NPR-B seem to be present in macrophages. The effect of ANP was mediated via the guanylate cyclase coupled NPR-A as shown by experiments employing stable cGMP analogs, the NPR-A antagonist HS-142-1, LY-83583, a cGMP inhibitor as well as C-ANF, a specific ligand of the NPR-C. Reduction of nitrite accumulation by ANP was highest when added simultaneously with LPS and abolished when added 12 h after LPS stimulation. In summary, ANP was shown to inhibit NO production of LPS-activated macrophages via cGMP.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Macrophages/metabolism , Nitric Oxide/biosynthesis , Aminoquinolines/pharmacology , Animals , Atrial Natriuretic Factor/antagonists & inhibitors , Base Sequence , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells , Cell Line , Cells, Cultured , Cyclic AMP/physiology , DNA/analysis , DNA/chemistry , DNA/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Macrophages/chemistry , Macrophages/drug effects , Mice , Natriuretic Peptide, C-Type , Oligonucleotides/analysis , Oligonucleotides/chemistry , Oligonucleotides/genetics , Peptide Fragments/pharmacology , Polymerase Chain Reaction , Polysaccharides/pharmacology , Proteins/pharmacology , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Atrial Natriuretic Factor/analysis , Receptors, Atrial Natriuretic Factor/genetics
19.
Eur J Pharmacol ; 336(2-3): 211-7, 1997 Oct 08.
Article in English | MEDLINE | ID: mdl-9384235

ABSTRACT

Inducible nitric oxide synthase dependent production of nitric oxide (NO) plays an important role in inflammation. We investigated whether pristimerin ((20alpha)-3-hydroxy-2-oxo-24-nor-friedela-1(10),3,5,7-te traen-carboxylic acid-(29)-methylester), an antitumoral, antimicrobial as well as anti-inflammatory plant compound, has an effect on the inducible NO synthase system in lipopolysaccharide-activated RAW 264.7 macrophages. Pristimerin dose dependently (IC50: 0.2-0.3 microM) reduces nitrite accumulation, a parameter for NO synthesis, in supernatants of lipopolysaccharide-stimulated (1 microg/ml, 20 h) macrophages. This effect correlates with a reduced inducible NO synthase enzyme activity measured by conversion of [3H]L-arginine to [3H]L-citrulline and significantly lower levels of enzyme protein (Western blotting) in homogenates of cells cotreated with lipopolysaccharide and pristimerin (12 h). Northern blot analysis and polymerase chain reaction (PCR) showed decreased inducible NO synthase mRNA levels in activated macrophages exposed to pristimerin (4 h). Electrophoretic mobility shift assay (EMSA) demonstrated a markedly reduced binding activity of nuclear factor-kappa B (NFkappaB) in nuclear extracts of pristimerin-treated cells. These results suggest that pristimerin inhibits the induction of inducible NO synthase by a mechanism which involves inhibition of NFkappaB activation. This feature of pristimerin is likely to contribute to its anti-inflammatory activity.


Subject(s)
Macrophages/drug effects , Nitric Oxide Synthase/biosynthesis , Triterpenes/pharmacology , Cell Line , Cyclooxygenase 2 , Enzyme Induction/drug effects , Isoenzymes/biosynthesis , Lipopolysaccharides , Macrophages/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Pentacyclic Triterpenes , Prostaglandin-Endoperoxide Synthases/biosynthesis , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , omega-N-Methylarginine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...