Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Biochem Mol Toxicol ; 26(2): 71-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22162043

ABSTRACT

Acetaminophen (APAP) is hepatotoxic and can cause toxicity in Jurkat T cells. p-Aminophenol (PAP), an industrial chemical and APAP metabolite, is nephrotoxic and hepatotoxic. Its potential toxicity in Jurkat T cells was investigated. PAP (10-250 µM) caused toxicity (decreased survival and increased LDH activity in incubation medium) and GSH depletion. At a concentration of 100 µM but not 250 µM, PAP increased DNA fragmentation. It decreased p-Akt levels (Elisa) and at higher concentrations decreased p-Akt expression (Western blotting). It had no effect on FasL expression. The cysteine precursor 2(RS)-n-propylthiazolidine-4(R)-carboxylic acid (250 µM) attenuated the PAP (100 µM)-induced decrease in viability and prevented GSH depletion and increased DNA fragmentation. It attenuated the PAP-induced decrease in p-Akt levels and protected against the decrease in p-Akt expression. The results demonstrate PAP-induced toxicity and suggest that it is due at least in part to apoptosis and involves GSH depletion and p-Akt inactivation.


Subject(s)
Aminophenols/toxicity , Cytoprotection , Oxidants/toxicity , T-Lymphocytes/drug effects , Thiazoles/pharmacology , Cell Survival/drug effects , Enzyme Activation , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Gene Expression/drug effects , Glutathione/metabolism , Humans , Jurkat Cells , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
2.
JPEN J Parenter Enteral Nutr ; 33(4): 380-9, 2009.
Article in English | MEDLINE | ID: mdl-19380752

ABSTRACT

BACKGROUND: Sepsis and septic shock syndrome are the leading causes of death in critically ill patients. Lipopolysaccharide (LPS) released by the colonic microorganisms may translocate across a compromised lumen, leading to upregulated reactive oxidative stress, inflammation, and sepsis. The authors examined an enteral formula high in cysteine (antioxidant precursor), omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and prebiotic fructooligosaccharides (FOS) against systemic inflammatory syndrome. METHODS: Rats were allocated to (1) standard soy-based diet high in cysteine and crude fiber and devoid of EPA-DHA (CHOW); (2) whey-peptide-based liquid diet high in cysteine, EPA-DHA, and FOS (CYSPUFA); or (3) casein-based liquid isonitrogenous diet, low in cysteine and devoid of EPA-DHA-FOS (CASN). Liquid diets provided 25% and CHOW, 23% of calories as protein. After 6 days on diets, rats received an intraperitoneal injection of LPS or saline. Animals gained weight on their respective diets and lost weight after LPS administration. The CYSPUFA group lost considerably less weight (vs CASN or CHOW, P < .05). Inflammatory cytokines significantly increased by 4 hours and subsided 18 hours after assault. The CASN group showed elevated liver enzyme alanine aminotransferase release from damaged hepatocytes and developed severe hepatic pathology with low hematocrit. The CHOW group developed more severe hepatic lesions compared with those on liquid diets. Concentration of liver enzyme and pathology were improved in rats receiving CYSPUFA. CONCLUSIONS: Data indicate that CYSPUFA, a diet rich in EPA-DHA-FOS, protects against LPS-induced systemic inflammatory responses and warrants clinical studies in critically ill patients.


Subject(s)
Cysteine/therapeutic use , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Enteral Nutrition/methods , Oligosaccharides/therapeutic use , Sepsis/prevention & control , Alanine Transaminase/blood , Animals , Antioxidants/analysis , Critical Illness , Cysteine/blood , Cytokines/blood , Disease Models, Animal , Humans , Lipopolysaccharides , Liver/pathology , Male , Random Allocation , Rats , Rats, Wistar , Sepsis/blood , Sepsis/pathology , Shock, Septic/prevention & control
3.
Dig Dis Sci ; 53(11): 2980-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18373199

ABSTRACT

Acetaminophen (APAP) elicits hepatotoxicity via multifactorial pathways, including increased apoptosis, cyclooxygenase (Cox-2) generation, reactive metabolite release, and glutathione (GSH) depletion. We previously showed that mice that consumed different antioxidants in their diets were protected against APAP-induced hepatotoxicity. We therefore further investigated the mechanisms by which green-tea polyphenols (GrTP) protect against APAP-induced hepatic damage. Mice were administered a diet supplemented with GrTP or vehicle for 5 consecutive days followed by intraperitoneal (IP) injection of a toxic dose of APAP or sham. APAP administration upregulated Cox-2 and B-cell lymphoma-2 (Bcl-2) production and had an effect, albeit minor, on Cox-1 and Fas expression in hepatic tissue. GrTP supplementation normalized APAP induced Cox-2 expression and Bcl-2 activation (P < 0.01), as evidenced by immunohistochemistry and Western blot analyses. Similarly, APAP administration elicited marked depletion (99%) in hepatic reduced GSH (rGSH) and endogenous S-adenosylmethionine (SAMe) concentrations (twofold) when compared with sham. APAP also caused severe centrilobular apoptosis and necrosis accompanied by leukocyte infiltration and marked elevations in the hepatic enzyme, alanine aminotransferase (ALT) released from damaged hepatocytes, and cytokine tumor necrosis factor alpha (TNF-alpha). GrTP improved hepatic histopathology (P < 0.01) and attenuated ALT activity (P < 0.05) and the depletion of rGSH (P < 0.05). In conclusion, GrTP supplementation attenuated hepatotoxicity by normalizing Cox-2 and Bcl-2 activation, suggesting a potential use for GrTP in treatng APAP toxicity.


Subject(s)
Acetaminophen/toxicity , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Flavonoids/pharmacology , Liver Diseases/metabolism , Phenols/pharmacology , Plant Extracts/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Alanine Transaminase/metabolism , Animals , Chemical and Drug Induced Liver Injury , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Glutathione/metabolism , Male , Mice , Mice, Inbred BALB C , Polyphenols , Tea/chemistry , Tumor Necrosis Factor-alpha/metabolism
4.
Dig Dis Sci ; 53(3): 767-76, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17710550

ABSTRACT

Methionine (Meth) is an essential amino acid involved in DNA methylation and glutathione biosynthesis. We examined the effect of Meth on the development of steatohepatitis. Rats were fed (five weeks) amino acid-based Meth-choline-sufficient (A-MCS) or total deficient (MCD) diets and gavaged daily (two weeks) with vehicle (B-vehicle/MCD), or Meth replacement (C-Meth/MCD). To assess the effect of short-term deficiency, after three weeks one MCS group was fed a deficient diet (D-MCS/MCD). Animals fed the deficient diet for two weeks lost (29%) weight and after five weeks weighed one third as much as those on the sufficient diet, and also developed anemia (P < 0.01). Hepatic transaminases progressively increased from two to five weeks (P < 0.01), leading to severe hepatic pathology. Meth administration normalized hematocrit, improved weight (P < 0.05), and suppressed abnormal enzymes activities (P < 0.01). Meth administration improved blood and hepatic glutathione (GSH), S-adenosylmethionine (SAMe), and hepatic lesions (P < 0.01). The deficient diet significantly upregulated proinflammatory and fibrotic genes, which was ameliorated by Meth administration. These data support a pivotal role for methionine in the pathogenesis of the dietary model of Meth-choline-deficient (MCD) steatohepatitis (NASH).


Subject(s)
Choline Deficiency/metabolism , Fatty Liver/metabolism , Hepatitis, Animal/metabolism , Liver/pathology , Methionine/deficiency , Animals , Choline Deficiency/pathology , Fatty Liver/pathology , Gene Expression , Hepatitis, Animal/pathology , Male , Rats , Rats, Sprague-Dawley
5.
J Nutr Biochem ; 19(3): 184-92, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17618106

ABSTRACT

4-Hydroxy-2-nonenal (HNE), the aldehydic product of lipid peroxidation, is associated with multiple immune dysfunctions, such as HIV and hepatitis C virus infection. HNE-induced immunosuppression could be due to a decrease in CD4+ T lymphocyte activation or proliferation. Glutathione (GSH) is the most abundant endogenous antioxidant in cells, and an adduct between HNE and GSH has been suggested to be a marker of oxidative stress. Our earlier studies showed that HNE induced cytotoxicity and Akt inactivation, which led to the enhancement of FasL expression and concomitantly decreased cellular FLICE-like inhibitory protein (c-FLIP(S)) levels. In this study, we found that HNE caused intracellular GSH depletion in Jurkat T cells, and we further investigated the role of 2(RS)-n-propylthiazolidine-4(R)-carboxylic acid (PTCA), a GSH prodrug, in attenuating HNE-induced cytotoxicity in CD4+ T lymphocytes. The results show that PTCA protected against HNE-induced apoptosis and depletion of intracellular GSH. PTCA also suppressed FasL expression through increasing levels of Akt kinase as well as antiapoptotic c-FLIP(S) and decreasing the activation of type 2 protein serine/threonine phosphatase. Taken together, these data demonstrate a novel correlation between GSH levels and Akt activation in T lymphocyte survival, which involves FasL down-regulation and c-FLIP(S) expression through increasing intracellular GSH levels. This suggests that PTCA could potentially be used in the treatment of oxidative stress-induced immunosuppressive diseases.


Subject(s)
Aldehydes/pharmacology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , Thiazoles/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/analysis , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/cytology , Enzyme Activation/drug effects , Fas Ligand Protein/genetics , Glutathione/analysis , Humans , Jurkat Cells , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/analysis , Signal Transduction/drug effects , fas Receptor/physiology
6.
Toxicol Appl Pharmacol ; 226(2): 128-39, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17919673

ABSTRACT

Exposure to arsenic via drinking water is a serious health concern in the US. Whereas studies have identified arsenic alone as an independent risk factor for liver disease, concentrations of arsenic required to damage this organ are generally higher than found in the US water supply. The purpose of the current study was to test the hypothesis that arsenic (at subhepatotoxic doses) may also sensitize the liver to a second hepatotoxin. To test this hypothesis, the effect of chronic exposure to arsenic on liver damage caused by acute lipopolysaccharide (LPS) was determined in mice. Male C57Bl/6J mice (4-6 weeks) were exposed to arsenic (49 ppm as sodium arsenite in drinking water). After 7 months of exposure, animals were injected with LPS (10 mg/kg i.p.) and sacrificed 24 h later. Arsenic alone caused no overt hepatotoxicity, as determined by plasma enzymes and histology. In contrast, arsenic exposure dramatically enhanced liver damage caused by LPS, increasing the number and size of necroinflammatory foci. This effect of arsenic was coupled with increases in indices of oxidative stress (4-HNE adducts, depletion of GSH and methionine pools). The number of apoptotic (TUNEL) hepatocytes was similar in the LPS and arsenic/LPS groups. In contrast, arsenic pre-exposure blunted the increase in proliferating (PCNA) hepatocytes caused by LPS; this change in the balance between cell death and proliferation was coupled with a robust loss of liver weight in the arsenic/LPS compared to the LPS alone group. The impairment of proliferation after LPS caused by arsenic was also coupled with alterations in the expression of key mediators of cell cycle progression (p27, p21, CDK6 and Cyclin D1). Taken together, these results suggest that arsenic, at doses that are not overtly hepatotoxic per se, significantly enhances LPS-induced liver injury. These results further suggest that arsenic levels in the drinking water may be a risk modifier for the development of chronic liver diseases.


Subject(s)
Arsenites/toxicity , Chemical and Drug Induced Liver Injury/etiology , Lipopolysaccharides/toxicity , Liver/drug effects , Sodium Compounds/toxicity , Animals , Apoptosis , Arsenites/administration & dosage , Body Weight/drug effects , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Drug Synergism , Hepatocytes/drug effects , Hepatocytes/pathology , Lipid Metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Sodium Compounds/administration & dosage
7.
Transl Res ; 150(2): 122-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17656332

ABSTRACT

Oxidant-mediated injury plays an important role in the pathophysiology of inflammatory bowel disease (IBD). Recently, antioxidants were shown to modulate colitis in mice. In this study, the protective effects of L-cysteine and glutathione (GSH) prodrugs are further evaluated against progression of colitis in a murine model. ICR mice were fed compounds incorporated into chow as follows: Group (A) received chow supplemented with vehicle. Group (B) was provided 2-(RS)-n-propylthiazolidine-4(R)-carboxylic-acid (PTCA), a cysteine prodrug. Group (C) received D-ribose-L-cysteine (RibCys), another cysteine prodrug that releases L-cysteine. Group (D) was fed L-cysteine-glutathione mixed sulfide (CySSG), a ubiquitous GSH derivative present in mammalian cells. After 3 days, the animals were further provided with normal drinking water or water supplemented with dextran sodium sulfate (DSS). Mice administered DSS developed severe colitis and suffered weight loss. Colonic lesions significantly improved in animals treated with PTCA and RibCys and, to a lesser extent, with CySSG therapy. Hepatic GSH levels were depleted in colitis animals (control vs DSS, P < 0.001), and normalized with prodrug therapies (control vs treatments, P > 0.05). Protein expressions of serum amyloid A and inflammatory cytokines [interleukin (IL)-6, IL-12, tumor necrosis factor-alpha (TNF-alpha), osteopontin (OPN)] were significantly increased in colitis animals and improved with therapies. Immunohistochemistry and Western blot analyses showed significant upregulation of the macrophage-specific markers, COX-2 and CD68, which suggests macrophage activation and infiltration in the colonic lamina propria in colitis animals. These abnormalities were attenuated in prodrug-treated mice. In conclusion, these data strongly support the novel action of the PTCA against colitis, which further supports a possible therapeutic application for IBD patients.


Subject(s)
Colitis/drug therapy , Cysteine/analogs & derivatives , Glutathione/analogs & derivatives , Thiazoles/administration & dosage , Thiazolidines/administration & dosage , Animals , Colon/pathology , Cysteine/administration & dosage , Cysteine/therapeutic use , Dextran Sulfate , Drug Delivery Systems , Glutathione/administration & dosage , Glutathione/therapeutic use , Interleukin-12/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred ICR , Sucrose , Thiazoles/therapeutic use , Thiazolidines/therapeutic use , Up-Regulation
8.
J Biochem Mol Toxicol ; 20(1): 39-47, 2006.
Article in English | MEDLINE | ID: mdl-16498637

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver (NAFL) and steatohepatitis (NASH) may accompany obesity, diabetes, parenteral nutrition, jejeuno-ileal bypass, and chronic inflammatory bowel disease. Currently there is no FDA approved and effective therapy available. We investigated the potential efficacy of those agents that stimulate glutathione (GSH) biosynthesis on the development of experimental steatohepatitis. Rats fed (ad libitum) amino acid based methionine-choline deficient (MCD) diet were further gavaged with (1) vehicle (MCD), (2) S-adenosylmethionine (SAMe), or (3) 2(RS)-n-propylthiazolidine-4(R)-carboxylic acid (PTCA). RESULTS: MCD diet significantly reduced hematocrit, and this abnormality improved in the treated groups (p < 0.01). Serum transaminases were considerably elevated (AST: 5.8-fold; ALT: 3.22-fold) in MCD rats. However, administration of GSH-enhancing agents significantly suppressed these abnormal enzyme activities. MCD rats developed severe liver pathology manifested by fatty degeneration, inflammation, and necrosis, which significantly improved with therapy. Blood levels of GSH were significantly depleted in MCD rats but normalized in the treated groups. Finally, RT-PCR measurements showed a significant upregulation of genes involved in tissue remodeling and fibrosis (matrix metalloproteinases, collagen-alpha1), suppressor of cytokines signaling1, and the inflammatory cytokines (IL-1beta, IL-6, TNF-alpha, and TGF-beta) in the livers of rats fed MCD. GSH-enhancing therapies significantly attenuated the expression of deleterious proinflammatory and fibrogenic genes in this dietary model. This is the first report that oral administration of SAMe and PTCA provide protection against liver injury in this model and suggests therapeutic applications of these compounds in NASH patients.


Subject(s)
Fatty Liver/diet therapy , Food, Formulated , Animals , Choline/administration & dosage , Cytokines/biosynthesis , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/pathology , Inflammation/blood , Inflammation/diet therapy , Inflammation/pathology , Lipotropic Agents/administration & dosage , Male , Methionine/administration & dosage , Rats , Rats, Sprague-Dawley , S-Adenosylmethionine/administration & dosage , Thiazoles/administration & dosage , Transaminases/blood
9.
J Nutr Biochem ; 16(5): 297-304, 2005 May.
Article in English | MEDLINE | ID: mdl-15866230

ABSTRACT

Reactive oxygen species (ROS) are increased in inflammatory bowel disease (IBD) and have been implicated as mediators of intestinal inflammation. We investigated the hypothesis that antioxidants with diverse properties attenuate disease progression in a murine dextran sodium sulfate (DSS)-induced colitis model. These antioxidants were (A) S-adenosylmethionine, a glutathione (GSH) precursor; (B) green tea polyphenols, a well-known antioxidant; and (C) 2(R,S)-n-propylthiazolidine-4(R)-carboxylic acid (PTCA), a cysteine prodrug, involved in GSH biosynthesis. BALB/c mice were divided into four groups and provided with the above mentioned antioxidants or the vehicle incorporated into chow. The animals were further divided into two subgroups and given normal drinking water (control) or water supplemented with DSS (to induce colitis), and the progression of the disease was studied. DSS-treated mice developed severe colitis as shown by bloody diarrhea, weight loss and pathological involvement (P<.001). However, all the antioxidants significantly improved diarrhea and colon lesions (P<.01), and increased body weights (P<.05). Hematocrits were significantly less affected in DSS-treated animals receiving antioxidants (P<.01). Colon lengths were significantly decreased due to mucosal inflammation in DSS-treated animals, but antioxidant therapy normalized this pathological finding (P<.001). The blood level of reduced GSH was decreased in DSS-treated mice (P<.05) and returned to normal when treated with antioxidants. Serum amyloid A (acute phase protein; P=.0015) and tumor necrosis factor-alpha (TNF-alpha; pro-inflammatory cytokine; P<.01) were significantly increased in DSS-treated animals (161+/-40 pg/ml) and improved with antioxidant treatment (P<.01). Finally, actin cytoskeleton was distorted and fragmented in the mucosa of DSS-treated mice and improved with antioxidant therapy. In conclusion, three structurally dissimilar antioxidants provided protection against DSS-induced colitis in this murine model, supporting a possible role for antioxidant therapy in IBD patients.


Subject(s)
Antioxidants/therapeutic use , Colitis/drug therapy , Animals , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Flavonoids/therapeutic use , Mice , Oxidative Stress/drug effects , Phenols/therapeutic use , Polyphenols , S-Adenosylmethionine/therapeutic use , Thiazoles/therapeutic use
10.
J Biochem Mol Toxicol ; 18(3): 154-61, 2004.
Article in English | MEDLINE | ID: mdl-15252872

ABSTRACT

p-Aminophenol (PAP) is a widely used industrial chemical and a known nephrotoxin. Recently, it was found to also cause hepatotoxicity and glutathione (GSH) depletion in mice. The exact mechanism of liver toxicity is not known. The aims of this study were to determine whether PAP can cause acute hepatotoxicity in hamsters and to further investigate the role of GSH in PAP-induced toxicity. PAP was administered ip to hamsters in doses of 200-800 mg/kg. Liver damage at 24 h after PAP administration was assessed by elevations in plasma enzyme activities and histopathologic examination. GSH and cysteine (Cys) levels in liver at 4 h were determined by HPLC. PAP decreased hepatic GSH concentration to 8% and Cys to 30% of vehicle control values. It increased plasma glutamic pyruvic transaminase (GPT) activity by 47-fold and sorbitol dehydrogenase (SDH) activity by 113-fold. PAP also caused severe centrilobular hepatocellular necrosis. 2(RS)-n-Propylthiazolidine-4(R)-carboxylic acid (PTCA), a Cys precursor, attenuated the PAP-induced decreases in hepatic sulfhydryl levels; GSH and Cys were 39% and 78% of vehicle controls, respectively. PTCA also attenuated the PAP-induced elevations in plasma enzyme activities and hepatic necrosis. It was concluded that PAP hepatotoxicity is associated with depletion of hepatic GSH and can be prevented by PTCA.


Subject(s)
Aminophenols/toxicity , Drug-Related Side Effects and Adverse Reactions/chemically induced , Glutathione/metabolism , Liver/drug effects , Alanine Transaminase/blood , Alanine Transaminase/drug effects , Animals , Chromatography, High Pressure Liquid , Cricetinae , Cysteine/analysis , Cysteine/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Kinetics , L-Iditol 2-Dehydrogenase/blood , L-Iditol 2-Dehydrogenase/drug effects , Liver/metabolism , Liver/pathology , Male , Methemoglobinemia/chemically induced , Protective Agents/pharmacology , Thiazoles/pharmacology
11.
J Am Coll Nutr ; 23(3): 220-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15190046

ABSTRACT

OBJECTIVE: Dietary, environmental and genetic events may influence host susceptibility to inflammatory bowel diseases (IBD). Transforming growth factor beta 2 (TGF-beta 2), a multifunctional polypeptide (cytokine) present in human and bovine milk, plays a critical role in the development of tolerance, the prevention of autoimmunity, and in anti-inflammatory responses. TGF-beta 2 is a potent inhibitor of intestinal epithelial cell (IEC) growth and stimulates IEC differentiation. The objective of this study was to determine whether a diet containing TGF-beta 2 modulates intestinal injury and immune responses in an Interleukin-10 knockout (IL-10-/-) mouse model of IBD. METHODS: Five-week-old IL-10-/- mice (in BALB/c background) reared in our transgenic facility were fed either an enteral diet (Diet-A) containing TGF-beta 2 or a control enteral diet (Diet-B) not rich in TGF-beta 2. Mice were weighed weekly, monitored for illness and euthanized after eight weeks on the diet. RESULTS: Final weights were 28 +/- 1.2 g (58.2% gain) for Diet-A mice and 23 +/- 1.6 g (32.9% gain) for Diet-B mice (p = 0.0194). The hematocrits were 48.3% for Diet-A compared to 42% for Diet-B mice (p = 0.0021). Mice on Diet-A had significantly lower serum TNF-alpha concentrations. Forty-four percent of mice on Diet-B developed severe diarrhea and rectal prolapse compared with none on Diet-A. Evaluation of intestinal pathology (score 0-4) revealed that animals fed Diet-A had a score of 2.1 +/- 0.4 compared to 3.2 +/- 0.36 in the Diet-B group (p = 0.040). The acute phase protein, serum amyloid A (SAA), was 3.8 times higher in the Diet-B group (p = 0.0038). CONCLUSIONS: IL-10-/- mice fed a TGF-beta 2 containing diet gained more weight, did not develop diarrhea or prolapse, had lower pathological scores, and lower SAAs. These data further support the use of TGF-beta 2 containing enteral diets as one mode of therapy for Crohn's disease.


Subject(s)
Immunosuppressive Agents/therapeutic use , Inflammatory Bowel Diseases/diet therapy , Nutritional Support/methods , Transforming Growth Factor beta/therapeutic use , Animals , Antioxidants/metabolism , Apolipoproteins/drug effects , Body Weight/drug effects , Cytokines/blood , Cytokines/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Interleukin-12/blood , Interleukin-18/blood , Mice , Mice, Inbred BALB C , Serum Amyloid A Protein/drug effects , Transforming Growth Factor beta2 , Treatment Outcome , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
J Biochem Mol Toxicol ; 18(6): 361-8, 2004.
Article in English | MEDLINE | ID: mdl-15674847

ABSTRACT

The reactive oxygen species-sensitive transcription nuclear factor-kappaB (NF-kappaB) plays a pivotal role in the development of acetaminophen (APAP) hepatotoxicity. We investigated the efficacy of a diverse series of antioxidants in preventing APAP-induced hepatotoxicity. BALB/c mice were divided into four groups and provided with antioxidants incorporated into chow as follows: (1) control diet; or diet supplemented with (2) S-adenosylmethionine (SAMe); (3) green tea polyphenols (GrTP); or (4) (RS)-n-propylthiazolidine-4(R)-carboxylic acid (PTCA). After 5 days on these diets, the animals were further subdivided into (A) given an IP injection with APAP (750 mg/kg), or (B) kept as untreated controls. The animals were sacrificed at 0, 4 h, and 24 h following APAP administration. PAP/vehicle induced marked decreases in hepatic reduced glutathione (GSH) levels and endogenous SAMe concentrations (46%) when compared to controls. APAP also caused severe centrilobular necrosis and marked increase in serum enzyme ALT activity (38-fold). Oral administration of antioxidants significantly attenuated the APAP-induced liver damage and depletion of hepatic GSH. There were profound increases in serum TNF-alpha levels at 4 h following APAP administration in nonsupplemented compared to antioxidant-treated animals, but no significant differences noted after 24 h. Serum amyloid A increased in APAP-challenged mice irrespective of antioxidant treatment. Finally, hepatic SAMe concentrations were drastically decreased 24 h following APAP administration, and these decreases were attenuated by pretreatment with antioxidants. In conclusion, these orally administered antioxidants with dissimilar properties provided protection against liver damage, supporting the potential use of antioxidant therapy in patients with APAP toxicity. This is the first report that GrTP and oral administration of PTCA and SAMe can provide protection against APAP injury in this model.


Subject(s)
Acetaminophen/toxicity , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Animals , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chromatography, High Pressure Liquid , Glutathione/metabolism , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred BALB C
13.
J Leukoc Biol ; 71(1): 141-53, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11781390

ABSTRACT

CXC chemokines, which induce angiogenesis, have glutamine-leucine-arginine amino acid residues (ELR motif) in the amino terminus and bind CXCR2 and the Duffy antigen chemokine-binding protein. Duffy, a seven transmembrane protein that binds CXC and CC chemokines, has not been shown to couple to trimeric G proteins or to transduce intracellular signals, although it is highly expressed on red blood cells, endothelial cells undergoing neovascularization, and neuronal cells. The binding of chemokines by Duffy could modulate chemokine responses positively or negatively. Positive regulation could come through the presentation of chemokine to functional receptors, and negative regulation could come through Duffy competition with functional chemokine receptors for chemokine binding, thus serving as a decoy receptor. To determine whether Duffy has a role in angiogenesis and/or maintenance of homeostasis, we developed transgenic mice expressing mDuffy under the control of the preproendothelin promoter/enhancer (PPEP), which directs expression of the transgene to the endothelium. Two PPEP-mDuffy-transgenic founders were identified, and expression of the transgene in the endothelium was verified by Northern blot, RT-PCR, and immunostaining of tissues. The phenotype of the mice carrying the transgene appeared normal by all visual parameters. However, careful comparison of transgenic and nontransgenic mice revealed two phenotypic differences: mDuffy-transgenic mice exhibited a diminished angiogenic response to MIP-2 in the corneal micropocket assay, and mDuffy-transgenic mice exhibited enhanced hepatocellular toxicity and necrosis as compared with nontransgenic littermates in response to overdose of acetaminophen (APAP; 400 mg/kg body weight). Morover, APAP treatment was lethal in 50% of the mDuffy-transgenic mice 24 h post challenge, and 100% of the nontransgenic littermates survived this treatment at the 24 h time point. Our data suggest that enhanced expression of mDuffy on endothelial cells can lead to impaired angiogenic response to chemokines and impaired maintenance of homeostasis in response to toxic stresses.


Subject(s)
Antigens, Protozoan , Carrier Proteins/genetics , Chemokines, CXC/physiology , Homeostasis/genetics , Neovascularization, Pathologic/genetics , Protozoan Proteins , Receptors, Cell Surface/genetics , Stress, Physiological/physiopathology , Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Animals , Carrier Proteins/physiology , Endothelium, Corneal/physiology , Liver/pathology , Liver/physiopathology , Mice , Mice, Transgenic , Necrosis , Receptors, Cell Surface/physiology , Receptors, Interleukin-8B/physiology , Stress, Physiological/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...