Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(3): 244-248, 2024 Mar 12.
Article in Chinese | MEDLINE | ID: mdl-38448176

ABSTRACT

Following the global outbreak of COVID-19, many patients have suffered from multi-system complications and long-term sequelae caused by the virus. Diaphragm dysfunction is an obscure post-COVID-19 symptom. Although a few cases of diaphragm dysfunction caused by COVID-19 infection have been reported abroad, there are no relevant reports in China. Herein, we present two cases of patients with respiratory distress after COVID-19 infection. On admission, dynamic chest radiographs revealed diaphragm dysfunction in these patients. Further investigations including diaphragm ultrasound, neurophysiological examinations, transdiaphragmatic pressure measurements cranial MRI, and antibody testing for autoimmune diseases, were conducted. The final diagnoses were severe myasthenia gravis induced by COVID-19 infection and diaphragmatic nerve and muscle involvement caused by COVID-19 infection. Both patients showed improvement in symptoms after treatment. Therefore, we summarized our case, with a review of the relevant literature to improve the understanding of the disease and to provide clinical evidence for future diagnosis and treatment.


Subject(s)
Autoimmune Diseases , COVID-19 , Humans , Diaphragm , Thorax , China
2.
Toxicol In Vitro ; 70: 105036, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33164849

ABSTRACT

Vanadium is considered as "possibly carcinogenic to humans" (V2O5, IARC Group 2B), yet uncertainties persist related to the toxicity mechanisms of the multiple forms of vanadium. Exposure to vanadium often co-occurs with other metals or with organic compounds that can be transformed by cytochrome p450 (CYP) enzymes into DNA-reactive carcinogens. Therefore, effects of a soluble form of vanadium (sodium metavanadate, NaVO3) and aflatoxin-B1 (AFB1) were tested separately and together, for induction of CYP activities, DNA damage (γH2AX and DNA alkaline unwinding assays), and DNA methylation changes (global genome and DNA repeats) in HepaRG or HepG2 liver cell lines. NaVO3 (≥ 2.3 µM) reduced CYP1A1 and CYP3A4 activities and induced DNA damage, butcaused important cell proliferation only in HepaRG cells. As a binary mixture, NaVO3 did not modify the effects of AFB1. There was no reproducible effect of NaVO3 (<21 µM) on DNA methylation in AluYb8, satellite-α, satellite-2, and by the luminometric methylation assay, but DNA methylation flow-cytometry signals in HepG2 cells (25-50 µM) increased at the G1 and G2 cell cycle phases. In conclusion, cell lines responded differently to NaVO3 supporting the importance of investigating more than one cell line, and a carcinogenic role of NaVO3 might reside at low concentrations by stimulating the proliferation of tumorigenic cells.


Subject(s)
Aflatoxin B1/toxicity , Cytochrome P-450 Enzyme System/metabolism , DNA Damage , DNA Methylation/drug effects , Liver/cytology , Vanadates/toxicity , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Humans , Microsomes, Liver/metabolism
3.
J Toxicol Environ Health A ; 82(1): 1-27, 2019.
Article in English | MEDLINE | ID: mdl-30744511

ABSTRACT

The notion that adverse health effects produced by exposure to environmental contaminants (EC) may be modulated by the presence of non-chemical stressors is gaining attention. Previously, our lab demonstrated that cross-fostering (adoption of a litter at birth) acted as a non-chemical stressor that amplified the influence of developmental exposure to EC on the glucocorticoid stress-response in adult rats. Using liver from the same rats, the aim of the current study was to investigate whether cross-fostering might also modulate EC-induced alterations in hepatic gene expression profiles. During pregnancy and nursing, Sprague-Dawley dams were fed cookies laced with corn oil (control, C) or a chemical mixture (M) composed of polychlorinated biphenyls (PCB), organochlorine pesticides (OCP), and methylmercury (MeHg), at 1 mg/kg/day. This mixture simulated the contaminant profile reported in maternal human blood. At birth, some control and M treated litters were cross-fostered to form two additional groups with different biological/nursing mothers (CC and MM). The hepatic transcriptome was analyzed by DNA microarray in male offspring at postnatal days 21 and 78-86. Mixture exposure altered the expression of detoxification and energy metabolism genes in both age groups, but with different sets of genes affected at day 21 and 78-86. Cross-fostering modulated the effects of M on gene expression pattern (MM vs M), as well as expression of energy metabolism genes between control groups (CC vs C). In conclusion, while describing short and long-term effects of developmental exposure to EC on hepatic transcriptomes, these cross-fostering results further support the consideration of non-chemical stressors in EC risk assessments.


Subject(s)
Environmental Pollutants/adverse effects , Gene Expression/genetics , Hydrocarbons, Chlorinated/adverse effects , Liver/drug effects , Methylmercury Compounds/adverse effects , Polychlorinated Biphenyls/adverse effects , Animals , Fetus/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley
4.
Toxicology ; 308: 20-33, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23537661

ABSTRACT

Perinatal events can reprogram the hypothalamo-pituitary-adrenal axis for the entire lifespan leading to abnormal glucocorticoid stress-response (GSR) in adulthood: a phenomenon reported to be mediated by changes in DNA methylation of the glucocorticoid receptor (GR) gene promoter. We examined whether in utero and/or lactational exposure to mixtures of environmental contaminants can also induce abnormal GSR during adulthood. The experiment included nine treatment groups. From gestation day (GD) 0 until postnatal day (PND) 20, dams were fed daily with a cookie laced with corn oil (control) or a chemical mixture (M) [polychlorinated biphenyls (PCBs), organochlorine pesticides, and methylmercury] at 0.5 or 1.0mg/kg/day (0.5M, and M). At birth, some control (C) and M litters were cross-fostered to create four groups with the following in utero/postnatal exposure: C/C, M/C, C/M, M/M. Other dams received 1.8ng/kg/day of a mixture of aryl hydrocarbon receptor (AhR) agonists (non-ortho PCBs, PC-dibenzodioxins and PC-dibenzofurans) without or with 0.5M (0.5MAhR). In adult male offspring the abundance of GR in treated groups was not different from the control, but the AhR and M groups were significantly different from each other with opposite effects in the hippocampus and liver. There was no change in DNA methylation of the GR promoter (exon-17 and -110). Abnormal GSRs were detected in the AhR, 0.5MAhR, CM, and MM groups. The literature associates abnormal GSR with metabolic and mental health impairments, thus these results support further investigation of the influence of developmental exposure to environmental contaminants and predisposition to stress-induced diseases.


Subject(s)
DNA Methylation/physiology , Environmental Pollutants/toxicity , Glucocorticoids/metabolism , Lactation/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptors, Glucocorticoid/metabolism , Animals , DNA Methylation/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , Lactation/drug effects , Liver/drug effects , Liver/metabolism , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/genetics
5.
Int J Toxicol ; 31(5): 454-66, 2012.
Article in English | MEDLINE | ID: mdl-22914890

ABSTRACT

Exposure to environmental contaminants induces the activation of cytochrome P450s (CYP) which lead to the hydroxylation of contaminants and endogenous hormones such as estrogens. The hydroxylation of estrogens forms catecholestrogens (CEs), one of them being the mutagenic 4-hydroxyestradiol-17ß (4-OH-E2). Catecholestrogens are transformed by catechol-o-methyltransferases (COMTs) into nonreactive methoxyestrogens. To investigate the hepatic metabolism of estradiol-17ß in female offspring at postnatal day (PND) 21, pregnant rats were dosed daily from gestation day 1 until PND 21 with 2 dose levels of organochlorine pesticides (OCPs; 0.019 or 1.9 mg/kg per d), methylmercury (MeHg; 0.02 or 2 mg/kg per d), polychlorinated biphenyls (PCBs; 0.011 or 1.1 mg/kg per d), or a mixture (M; 0.05 or 5 mg/kg per d) including all 3 groups of chemicals. Concentrations of organochlorines in the mixture M were based on their proportions in serum of the Canadian Arctic population. The messenger RNA (mRNA) expressions of CYP and COMT were analyzed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). High-performance thin layer chromatography and phosphor imaging were used to measure the transformation of (14)C substrates into estrogen metabolites. The low-dose treatments or the MeHg groups had no effect. The high-dose OCP, PCB, and M group increased the production of 2-OH-E2 and 6α-OH-E2, while only the PCB and M groups increased the 2-OH-CE/methoxyestrogen ratio. In all groups, the cytosolic COMT activity exceeded the microsomal production rate of 4-OH-E2. Although the M treatment included the PCB and OCP mixtures, it did not modify the estrogen metabolism more than did the PCB mixture alone. This endocrine disruption information contributes to our understanding of chemical interactions in the toxicology of chemical mixtures.


Subject(s)
Endocrine Disruptors/toxicity , Environmental Pollutants/toxicity , Estradiol/metabolism , Hydrocarbons, Chlorinated/toxicity , Methylmercury Compounds/toxicity , Pesticides/toxicity , Animals , Catechol O-Methyltransferase/genetics , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Liver/drug effects , Liver/metabolism , Maternal-Fetal Exchange , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley
6.
Transplant Proc ; 42(5): 1830-4, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20620533

ABSTRACT

We used in situ perfusion and a multiple-organ harvesting technique to collect islets from adult pig pancreata. The tissues were digested with collagenase P followed by purification in a lympholyte discontinuous gradient using a COBE2991 cell separator. The yield and purity of isolated islets were evaluated with a light microscope after dithizone (DTZ) staining. Islet function was assessed using an in vitro insulin release assay. The results showed that before purification 275,000 +/- 20,895 islet equivalents (IEQ) were obtained from 1 digested pancreas. After purification with gradient centrifugation, the islet yield was 230,350 +/- 26,679 IEQ/pancreas. Each gram of the purified pancreatic tissues yielded 2710 +/- 229 IEQ with an average purity of 50.2 +/- 2.0%. The purified islet cells responded to stimulation with high glucose concentrations (16.7 mmol/L), namely, 4.74-fold greater than the insulin secretion with exposure to the basal level of glucose (3.3 mmol/L; P < .001). These results suggested that the established isolation method can be applied to large-scale purification of fully functional islets from pig pancreata.


Subject(s)
Islets of Langerhans/cytology , Animals , Cell Count , Cell Separation/methods , Cell Size , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/statistics & numerical data , Organ Size , Pancreas/anatomy & histology , Pancreas/cytology , Swine
7.
Oncogene ; 25(44): 5960-8, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16652148

ABSTRACT

The neurofibromatosis type 2 NF2 gene product, merlin, is a tumor suppressor frequently inactivated in malignant mesothelioma (MM). To investigate a possible correlation between merlin inactivation and MM invasiveness, we restored merlin expression in NF2-deficient MM cells. Re-expression of merlin markedly inhibited cell motility, spreading and invasiveness, properties connected with the malignant phenotype of MM cells. To test directly whether merlin inactivation promotes invasion in a nonmalignant system, we used small interfering RNA to silence Nf2 in mouse embryonic fibroblasts (MEFs) and found that downregulation of merlin resulted in enhanced cell spreading and invasion. To delineate signaling events connected with this phenotype, we investigated the effect of merlin expression on focal adhesion kinase (FAK), a key component of cellular pathways affecting migration and invasion. Expression of merlin attenuated FAK phosphorylation at the critical phosphorylation site Tyr397 and disrupted the interaction of FAK with its binding partners Src and p85, the regulatory subunit of phosphatidylinositol-3-kinase. In addition, NF2-null MM cells stably overexpressing FAK showed increased invasiveness, which decreased significantly when merlin expression was restored. Collectively, these findings suggest that merlin inactivation is a critical step in MM pathogenesis and is related, at least in part, with upregulation of FAK activity.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Mesothelioma/pathology , Mesothelioma/prevention & control , Neurofibromin 2/biosynthesis , Neurofibromin 2/genetics , Animals , Cell Line, Tumor , Cell Migration Inhibition , Cells, Cultured , Down-Regulation/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Mesothelioma/enzymology , Mice , Neoplasm Invasiveness/prevention & control , Neurofibromin 2/physiology , Signal Transduction/genetics
8.
Toxicol Sci ; 86(1): 175-84, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15858227

ABSTRACT

Non-ortho polychlorinated biphenyls (PCBs), polychlorinated dibenzodioxins (PCDDs), and polychlorinated dibenzofurans (PCDFs) are ubiquitous environmental contaminants that exert their toxicity mostly through activation of the aryl-hydrocarbon receptor (AhR), and are referred to as AhR agonists. The objective was to study, by real time reverse-transcriptase-polymerase chain reaction (RT-PCR), the effects of postnatal exposure to a reconstituted mixture of AhR agonists present in breast milk (3 non-ortho PCBs, 6 PCDDs, and 7 PCDFs, referred to here-in-after as AhRM) on mRNA expression of estrogen receptor (ERalpha), enzymes involved with the metabolism of estrogens [catechol-o-methyltransferase (Comt), cytochrome P450 (Cyp)1A1, 1B1 and 2B1], and DNA methyltransferase-1 (Dnmt1), in brain areas, liver and uterus of immature female rats. Neonates were exposed by gavage during postnatal day (PND) 1-20 with dosages equivalent to 1, 10, 100, and 1000 times the estimated average human exposure level, and were sacrificed at PND 21. None of the end points were affected in uterine cross-sections, or in samples of uterine tissue layers collected by laser capture microdissection. At 1000x, the AhRM reduced Dnmt1 mRNA abundance to 28% and 32% of control in the liver and hypothalamus, respectively. In the brain, Cyp1A1 was increased (409%) but ERalpha was reduced (66%). Similarly, mRNA abundance for Comt isoforms was reduced in the liver (45%) and brain areas (55-70%). AhRM at 100x, the lowest effective dose, exerted a 220% increase in brain cortex Comt [membrane bound (Mb)], a 219% increase in hepatic Cyp1B1, and a 63% decrease in hepatic Comt (soluble (S)+Mb). These results support the possibility that early exposure to environmental contaminants could lead to effects mediated by changes in DNA methylation and/or estrogen metabolism and signaling.


Subject(s)
Brain/drug effects , Liver/drug effects , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/agonists , Uterus/drug effects , Animals , Base Sequence , Brain/enzymology , Brain/metabolism , Catechol O-Methyltransferase/genetics , Cytochrome P-450 Enzyme System/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Primers , Female , Liver/enzymology , Liver/metabolism , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Uterus/enzymology , Uterus/metabolism
9.
Toxicol Appl Pharmacol ; 199(3): 344-53, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15364549

ABSTRACT

Previous studies in neonatal and adolescent rats as well as adrenalectomized rats have demonstrated that glucocorticoids regulate the expression of the rat NAD(P)H:quinone oxidoreductase gene (QOR). We used primary cultures of rat adult hepatocytes to document that added glucorticoids repress both the basal and 1,2-benzanthracene-induced expression of QOR mRNA by 65-70%. QOR enzyme activity and protein were concomitantly suppressed as well. The monotonic concentration response for repression of QOR gene products up to 100 microM DEX concentration demonstrated that the glucocorticoid receptor (GR) was most likely involved in this process. The lack of effect at higher concentration rules out a role for the Pregnane X receptor in this regulation by DEX. In addition, the anti-glucorticoid RU38486 blocked this negative regulation and the protein synthesis inhibitor cycloheximide had no effect on this repression process. Similar results of GR dependence were observed using a luciferase reporter construct containing the 5'-flanking region of the human QOR gene using HepG2 cells. Collectively, these results demonstrate that GR must directly participate in the negative regulation of QOR gene expression by dexamethasone and other glucocorticoids in vivo.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , 5' Flanking Region/genetics , Animals , Blotting, Northern , Cells, Cultured , Cycloheximide/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Dexamethasone/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Hormone Antagonists/pharmacology , Luciferases/biosynthesis , Luciferases/genetics , Male , Mifepristone/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Glucocorticoid/drug effects , Transfection
10.
Toxicol Sci ; 75(2): 468-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12883090

ABSTRACT

There are concerns that postnatal exposure to organochlorines present in breast milk could lead to adverse health effects. We reconstituted four mixtures of aryl-hydrocarbon receptor (AhR) agonists (3 non-ortho polychlorinated biphenyls [PCBs], 6 polychlorinated dibenzodioxins [PCDDs], 7 polychlorinated dibenzofurans [PCDFs], or all 16 chemicals together [referred to as AhRM]) based on their concentrations in breast milk, and examined their effects following exposure by gavage from day 1 until day 20 of age. Female neonates received dosages of AhRM equivalent to 1, 10, 100, or 1000 times the amount consumed by an infant over the first 24 days of life. Other groups received the PCBs, the PCDDs, or the PCDFs at the 1000x level. All rats were sacrificed at 21 days of age. Changes in ethoxyresorufin-o-deethylase hepatic activity, thymus and body weights, and serum thyroxin were linked to the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) toxic equivalents (TEQ) of the four mixtures (1000x-AhRM > PCDDs > PCBs > PCDFs). To test for AhRM antiestrogenicity, two additional groups received 1.5 microg/kg of 17alpha-ethynyl estradiol (EE) with or without the 1000x-AhRM. The AhRM had no effect on uterine weight or EE-stimulated uterine growth. The actions of the combined EE and AhRM treatments suggest additive effects in decreasing pentoxyresorufin-o-deethylase activity and spleen weight, but nonadditive/antagonistic effects on adrenal weight and serum thyroxin. In conclusion, (1) 10x-AhRM had no detectable effects, (2) TEQ values relate to observed toxicities, even when testing complex mixtures of AhR agonists, and (3) indications of tissue-specific additive and nonadditive/antagonistic effects, but no synergism, were observed when doses of AhRM were increased, or combined with EE.


Subject(s)
Benzofurans/toxicity , Polychlorinated Biphenyls/toxicity , Polychlorinated Dibenzodioxins/analogs & derivatives , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/agonists , Soil Pollutants/toxicity , Animals , Animals, Newborn , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzofurans/administration & dosage , Biological Assay , Dibenzofurans, Polychlorinated , Dose-Response Relationship, Drug , Drug Combinations , Female , Organ Size/drug effects , Polychlorinated Biphenyls/administration & dosage , Polychlorinated Dibenzodioxins/administration & dosage , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Soil Pollutants/administration & dosage , Thymus Gland/drug effects , Thymus Gland/pathology , Thyroxine/blood , Uterus/drug effects , Uterus/growth & development , Uterus/pathology
11.
Mol Pharmacol ; 60(3): 611-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502894

ABSTRACT

Glucocorticoids regulate the rat glutathione S-transferase A2 (GSTA2) gene in a biphasic manner in cultured hepatocytes that repress gene expression at low concentration (10--100 nM) but induce gene expression at high concentration (>1 microM). High concentrations of the glucocorticoid receptor (GR) antagonist RU38486 (5--10 microM) also induced the expression of GSTA2. These effects were reproduced in HepG2 cells transfected with a luciferase reporter containing 1.6 kilobase pairs of 5'-flanking sequence of GSTA2 and expression plasmids for either GR, pregnane X receptor (PXR) or a combination of both. Dexamethasone t-butylacetate (1 microM t-Bu-DEX) repressed gene expression between 60 to 75% when only GR was expressed. When PXR was expressed, both basal and t-Bu-DEX-dependent gene expression was increased over 2-fold, respectively. Biphasic regulation of gene expression was observed over a broad range of t-Bu-DEX concentrations when expression plasmids for both receptors were cotransfected. Other steroids of the pregnane class induced GSTA2 expression as expected for a PXR-dependent process. Because no canonical responsive element for the PXR-RXR alpha heterodimer was observed in the 5'-flanking region of the construct, deletion analysis was used to identify a pregnane responsive region between base pairs -700 and -683; this 20-bp region contains the antioxidant response element (ARE). When the ARE sequence was mutated, basal, t-butylhydroquinone- and 17 alpha-hydroxypregnenolone-inducible expression were all lost. These results suggest that PXR interacts with factors binding to the ARE to elicit the pregnane inductive response for GSTA2.


Subject(s)
Glucocorticoids/pharmacology , Glutathione Transferase/metabolism , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Animals , Antioxidants/pharmacology , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Gene Deletion , Genes, Reporter , Glutathione Transferase/drug effects , Glutathione Transferase/genetics , Hepatocytes/enzymology , Hormone Antagonists/pharmacology , Humans , Hydroquinones/pharmacology , Male , Mifepristone/pharmacology , Pregnane X Receptor , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
12.
Cancer Res ; 61(2): 589-93, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212254

ABSTRACT

Insulin-like growth factor I receptor (IGF-IR) is frequently overexpressed in several types of human malignancy and is associated with invasion and metastasis of tumor cells. Recently, IGF-IR expression was reported to be up-regulated in the human pancreatic cancer cell line PANC-1 when cells were stably transfected with active Src. The downstream targets of Src that lead to the up-regulation of IGF-IR expression were previously unknown. We demonstrate here that AKT regulates IGF-IR expression in PANC-1 and AsPC-1 cells. Cells transfected with active Src exhibited significantly more IGF-IR protein compared with vector-transfected cells. Overexpression of wild-type or constitutively active AKT (i.e., AKT1 or AKT2) also resulted in elevated IGF-IR expression. IGF-IR protein levels were higher in cells transfected with constitutively active AKT than in cells transfected with active Src. In vitro kinase assays showed that AKT kinases are activated by active Src and inhibited by dominant negative Src or the tumor suppressor PTEN. Furthermore, AKT-induced IGF-IR expression was down-regulated by dominant-negative Src or PTEN. In addition, cells transfected with activated AKT in the presence of IGF-I were shown to have enhanced invasiveness compared with control cells. These data provide evidence for a link between AKT signaling and the regulation of IGF-IR expression and demonstrate that active AKT promotes the invasiveness of pancreatic cancer cells through the up-regulation of IGF-IR expression.


Subject(s)
Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor, IGF Type 1/metabolism , Tumor Suppressor Proteins , Blotting, Western , DNA, Recombinant , Down-Regulation , Enzyme Activation , Humans , Neoplasm Invasiveness , PTEN Phosphohydrolase , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphoric Monoester Hydrolases/metabolism , Plasmids/genetics , Precipitin Tests , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transfection , Tumor Cells, Cultured , Up-Regulation , src-Family Kinases/metabolism
13.
Toxicol Sci ; 59(2): 226-30, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158715

ABSTRACT

2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is a heterocyclic amine carcinogen present in well-done meat. PhIP must undergo host-mediated bioactivation to exert its mutagenic and carcinogenic effects. Following N-hydroxylation, N-acetyltransferases catalyze the O-acetylation (activation) of N-hydroxy-PhIP to an electrophile causing DNA damage. A well-defined genetic polymorphism in N-acetyltransferase 2 (NAT2) activity exists in humans and the Syrian hamster. Since some human epidemiological studies suggest an association between acetylator genotype and cancer susceptibility in individuals who consume well done meats, this study was designed to investigate the specific role of acetylator genotype in PhIP-induced tumors using a Syrian hamster model congenic at the NAT2 locus. Following oral administration of PhIP to male rapid and slow acetylator Syrian hamsters, DNA adducts were identified in each tissue examined with levels in the relative order: pancreas > heart and urinary bladder > prostate, small intestine and transverse colon > ascending colon, liver, cecum, descending colon, and rectum. However, no tumors were observed in male rapid and slow acetylator congenic hamsters administered 11 oral doses of PhIP (75 mg/kg) and maintained on a high fat diet for one year.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Carcinogens/toxicity , DNA Adducts/drug effects , Imidazoles/toxicity , Acetylation , Animals , Animals, Congenic , Cricetinae , DNA/drug effects , DNA Adducts/analysis , Disease Models, Animal , Imidazoles/metabolism , Male , Mesocricetus , Polymorphism, Genetic
14.
J Biochem Mol Toxicol ; 15(1): 26-33, 2001.
Article in English | MEDLINE | ID: mdl-11170312

ABSTRACT

N-acetyltransferases (EC 2.3.1.5) catalyze O-acetylation of heterocyclic amine carcinogens to DNA-reactive electrophiles that bind and mutate DNA. An acetylation polymorphism exists in humans and Syrian hamsters regulated by N-acetyltransferase-2 (NAT2) genotype. Some human epidemiological studies suggest a role for NAT2 phenotype in predisposition to cancers related to heterocyclic amine exposures, including breast cancer. 2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) is a heterocyclic amine carcinogen prevalent in the human environment and induces a high incidence of mammary tumors in female rats. PhIP-induced carcinogenesis was examined in female rapid and slow acetylator Syrian hamsters congenic at the NAT2 locus. In both rapid and slow acetylators, PhIP-DNA adduct levels were highest in pancreas, lower in heart, small intestine, and colon, and lowest in mammary gland and liver. Metabolic activation of N-hydroxy-PhIP by O-acetyltransferase was highest in mammary epithelial cells, lower in liver and colon, and lowest in pancreas. Metabolic activation of N-hydroxy-PhIP by O-sulfotransferase was low in liver and colon and below the limit of detection in mammary epithelial cells and pancreas. Unlike the rat, PhIP did not induce breast or any other tumors in female rapid and slow acetylator congenic hamsters administered high-dose PhIP (10 doses of 75 mg/kg) and a high-fat diet.


Subject(s)
Carcinogens/toxicity , DNA Adducts/drug effects , Imidazoles/toxicity , Mammary Neoplasms, Experimental/chemically induced , Acetylation , Administration, Oral , Animals , Animals, Congenic , Arylamine N-Acetyltransferase/genetics , Carcinogens/administration & dosage , Cricetinae , DNA Adducts/metabolism , Disease Models, Animal , Female , Homozygote , Imidazoles/administration & dosage , Imidazoles/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mesocricetus , Tissue Distribution
15.
Proc Natl Acad Sci U S A ; 98(1): 247-52, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11134526

ABSTRACT

Hepatocyte growth factor (HGF) is a ligand of the receptor tyrosine kinase encoded by the c-Met protooncogene. HGF/Met signaling has multifunctional effects on various cell types. We sought to determine the role of HGF/Met in apoptosis and identify signal transducers involved in this process. In experiments with human SK-LMS-1 leiomyosarcoma cells, we show that the Akt kinase is activated by HGF in a time- and dose-dependent manner by phosphatidylinositol 3-kinase (PI3-kinase). Akt is also activated by active tumorigenic forms of Met, i.e., ligand-independent Tpr-Met, a truncated and constitutively dimerized form of Met, and a mutationally activated version of Met corresponding to that found in human hereditary papillary renal carcinoma. In NIH 3T3 cells transfected with wild-type Met, HGF inhibits apoptosis induced by serum starvation and UV irradiation. HGF-induced survival correlates with Akt activity and is inhibited by the specific PI3-kinase inhibitor LY294002, indicating that HGF inhibits cell death through the PI3-kinase/Akt signal transduction pathway. Furthermore, transiently transfected Tpr-Met activates Akt (both Akt1 and Akt2) and protects cells from apoptosis. Mitogen-activated protein kinase (MAPK) also is activated by HGF and rescues cells from apoptosis, although the cytoprotective effect is less marked than for PI3-kinase/Akt. Blocking MAPK with the specific MAPK kinase inhibitor PD098059 impairs the ability of HGF to promote cell survival. Similar results were obtained with NIH 3T3 cells expressing the fusion protein Trk-Met and stimulated with nerve growth factor, the Trk ligand. These results demonstrate that HGF/Met is capable of protecting cells from apoptosis by using both PI3-kinase/Akt and, to a lesser extent, MAPK pathways.


Subject(s)
Apoptosis/drug effects , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Chromones/pharmacology , Cytoprotection , Enzyme Activation/drug effects , Flavonoids/pharmacology , Humans , Ligands , Mice , Morpholines/pharmacology , Oncogene Proteins, Fusion/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-met/metabolism , Transfection , Tumor Cells, Cultured
16.
Cancer Epidemiol Biomarkers Prev ; 9(9): 905-10, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11008907

ABSTRACT

Heterocyclic amines found in well-done meat require host-mediated metabolic activation before initiating DNA mutations and tumors in target organs. Polymorphic N-acetyltransferase-2 (NAT2) catalyzes the activation of heterocyclic amines via O-acetylation, suggesting that NAT2 genotypes with high O-acetyltransferase activity (rapid/intermediate acetylator phenotype) increase the risk of breast cancer in women who consume well-done meat. To test this hypothesis, DNA samples and information on diet and other breast cancer risk factors were obtained from a nested case-control study of postmenopausal women. Twenty-seven NAT2 genotypes were determined and assigned to rapid, intermediate, or slow acetylator groups based on published characterizations of recombinant NAT2 allozymes. NAT2 genotype alone was not associated with breast cancer risk. A significant dose-response relationship was observed between breast cancer risk and consumption of well-done meat among women with the rapid/intermediate NAT2 genotype (trend test, P = 0.003) that was not evident among women with the slow acetylator genotype (trend test, P = 0.22). These results suggest an interaction between NAT2 genotype and meat doneness, although a test for multiplicative interaction was not statistically significant (P = 0.06). Among women with the rapid/intermediate NAT2 genotype, consumption of well-done meat was associated with a nearly 8-fold (odds ratio, 7.6; 95% confidence interval, 1.1-50.4) elevated breast cancer risk compared with those consuming rare or medium-done meats. These results are consistent with a role for O-acetylation in the activation of heterocyclic amine carcinogens and support the hypothesis that the NAT2 acetylation polymorphism is a breast cancer risk factor among postmenopausal women with high levels of heterocyclic amine exposure.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Meat/adverse effects , Acetylation , Aged , Amines/adverse effects , Biotransformation , Breast Neoplasms/epidemiology , Breast Neoplasms/etiology , Carcinogens , Case-Control Studies , Cooking , Female , Heterocyclic Compounds/adverse effects , Humans , Likelihood Functions , Logistic Models , Middle Aged , Odds Ratio , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Postmenopause/physiology , Risk Factors
17.
Biochem J ; 349(Pt 1): 141-51, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861222

ABSTRACT

Choline acetyltransferase synthesizes acetylcholine in cholinergic neurons and, in humans, may be produced in 82- and 69-kDa forms. In this study, recombinant choline acetyltransferase from baculovirus and bacterial expression systems was used to identify protein isoforms by two-dimensional SDS/PAGE and as substrate for protein kinases. Whereas hexa-histidine-tagged 82- and 69-kDa enzymes did not resolve as individual isoforms on two-dimensional gels, separation of wild-type choline acetyltransferase expressed in insect cells revealed at least nine isoforms for the 69-kDa enzyme and at least six isoforms for the 82-kDa enzyme. Non-phosphorylated wild-type choline acetyltransferase expressed in Escherichia coli yielded six (69 kDa) and four isoforms (82 kDa) respectively. Immunofluorescent labelling of insect cells expressing enzyme showed differential subcellular localization with the 69-kDa enzyme localized adjacent to plasma membrane and the 82-kDa enzyme being cytoplasmic at 24 h. By 64 h, the 69-kDa form was in cytoplasm and the 82-kDa form was only present in nucleus. Studies in vitro showed that recombinant 69-kDa enzyme was a substrate for protein kinase C (PKC), casein kinase II (CK2) and alpha-calcium/calmodulin-dependent protein kinase II (alpha-CaM kinase), but not for cAMP-dependent protein kinase (PKA); phosphorylation by PKC and CK2 enhanced enzyme activity. The 82-kDa enzyme was a substrate for PKC and CK2 but not for PKA or alpha-CaM kinase, with only PKC yielding increased enzyme activity. Dephosphorylation of both forms of enzyme by alkaline phosphatase decreased enzymic activity. These studies are of functional significance as they report for the first time that phosphorylation enhances choline acetyltransferase catalytic activity.


Subject(s)
Choline O-Acetyltransferase/biosynthesis , Choline O-Acetyltransferase/chemistry , Animals , Baculoviridae/metabolism , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Casein Kinase II , Catalysis , Cell Line , Cell Membrane/metabolism , Choline O-Acetyltransferase/isolation & purification , Choline O-Acetyltransferase/metabolism , Chromatography, Agarose , Cytoplasm/metabolism , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Humans , Immunohistochemistry , Insecta , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation , Protein Isoforms , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Time Factors
18.
Cancer Epidemiol Biomarkers Prev ; 9(1): 29-42, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10667461

ABSTRACT

The focus of this review is the molecular genetics, including consensus NAT1 and NAT2 nomenclature, and cancer epidemiology of the NAT1 and NAT2 acetylation polymorphisms. Two N-acetyltransferase isozymes, NAT1 and NAT2, are polymorphic and catalyze both N-acetylation (usually deactivation) and O-acetylation (usually activation) of aromatic and heterocyclic amine carcinogens. Epidemiological studies suggest that the NAT1 and NAT2 acetylation polymorphisms modify risk of developing urinary bladder, colorectal, breast, head and neck, lung, and possibly prostate cancers. Associations between slow NAT2 acetylator genotypes and urinary bladder cancer and between rapid NAT2 acetylator genotypes and colorectal cancer are the most consistently reported. The individual risks associated with NAT1 and/or NAT2 acetylator genotypes are small, but they increase when considered in conjunction with other susceptibility genes and/or aromatic and heterocyclic amine carcinogen exposures. Because of the relatively high frequency of some NAT1 and NAT2 genotypes in the population, the attributable cancer risk may be high. The effect of NAT1 and NAT2 genotype on cancer risk varies with organ site, probably reflecting tissue-specific expression of NAT1 and NAT2. Ethnic differences exist in NAT1 and NAT2 genotype frequencies that may be a factor in cancer incidence. Large-scale molecular epidemiological studies that investigate the role of NAT1 and NAT2 genotypes and/or phenotypes together with other genetic susceptibility gene polymorphisms and biomarkers of carcinogen exposure are necessary to expand our current understanding of the role of NAT1 and NAT2 acetylation polymorphisms in cancer risk.


Subject(s)
Arylamine N-Acetyltransferase/genetics , Isoenzymes/genetics , Polymorphism, Genetic/genetics , Acetylation , Biomarkers/analysis , Carcinogens/metabolism , Colonic Neoplasms/etiology , Ethnicity/genetics , Genetic Predisposition to Disease , Genotype , Humans , Incidence , Molecular Biology , Molecular Epidemiology , Phenotype , Rectal Neoplasms/etiology , Risk Factors , Terminology as Topic , Urinary Bladder Neoplasms/etiology
19.
Adv Exp Med Biol ; 463: 159-64, 1999.
Article in English | MEDLINE | ID: mdl-10352681

ABSTRACT

The expression of the aldehyde dehydrogenase 3 gene is known to be controlled by multiple regulatory processes. In liver, inducible expression appears to be mediated by two AhRE sequences which allow regulation of this gene by xenobiotic compounds which are ligands for the Ah receptor (Takimoto et al., 1994; this work). Constitutive expression of ALDH3 in tissues such as the cornea also involves the -3,500 region which contains an AhRE (Boesch et al., 1996; Boesch et al, 1998). However, the constellation of transcription factors which appear to interact with the AhRE in constitutively expressing corneal cells does not include either the Ah receptor nor the prototypical ARNT protein (Boesch et al., 1998). For both inducible and constitutive ALDH3 expression the more distal 5' flanking region sequences appear to interact with more proximal regulatory elements. Of particular interest is the region near -1 kb which includes the GC (-930 to -910) and cAMP (-1057 to -991) responsive elements as well as the 2 NF1 sites (-916 to -815), all of which appear to act as negative modulators of ALDH3 expression. A second putative ALDH3 negative regulatory region lies even more distal than -3,500 bp. To date, this region has been little studied, but appears to be involved in regulating both inducible and constitutive ALDH3 expression. This region may also be responsible for some of the tissue-specificity of ALDH3 expression. With respect to the work described here, in both isolated hepatocytes and HepG2 cells, no consistent negative regulation by glucocorticoids was observed in the basal expression of ALDH3. This indicates that the mechanism of GC-mediated negative regulation involves direct interference with ALDH3 gene activation mediated by the Ah receptor. Our results suggest a complex interplay between multiple transcription factors, including the GC and Ah receptors, regulates the hepatic expression of the ALDH3 gene. Active recruitment of transcription factors needed for gene transactivation, amelioration of the actions of negative regulatory trans-acting factors or cis-acting elements and/or chromatin remodeling may be required for achieve proper regulation of the aldehyde dehydrogenase 3 gene.


Subject(s)
Aldehyde Dehydrogenase/genetics , Dexamethasone/metabolism , Gene Expression Regulation, Enzymologic , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Dexamethasone/pharmacology , Genes, Reporter , Glucocorticoids/pharmacology , Liver/cytology , Promoter Regions, Genetic , Rats , Transcriptional Activation , Transfection , Tumor Cells, Cultured
20.
Mol Pharmacol ; 55(4): 649-57, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10101022

ABSTRACT

Glucocorticoids repressed the polycyclic aromatic hydrocarbon-dependent induction of Class 3 aldehyde dehydrogenase (ALDH3) enzyme activity and mRNA levels in isolated rat hepatocytes by more than 50 to 80%, with a concentration-dependence consistent with the involvement of the glucocorticoid receptor (GR). No consistent effect on the low basal transcription rate was observed. This effect of glucocorticoids (GC) on polycyclic aromatic hydrocarbon induction was effectively antagonized at the mRNA and protein level by the GR antagonist RU38486. The response was cycloheximide-sensitive, because the protein synthesis inhibitor caused a GC-dependent superinduction of ALDH3 mRNA levels. This suggests that the effects of GC on this gene are complex and both positive and negative gene regulation is possible. The GC-response was recapitulated in HepG2 cells using transient transfection experiments with CAT reporter constructs containing 3.5 kb of 5'-flanking region from ALDH3. This ligand-dependent response was also observed when a chimeric GR (GR DNA-binding domain and peroxisome proliferator-activated receptor ligand-binding domain) was used in place of GR in the presence of the peroxisome proliferator, nafenopin. A putative palindromic glucocorticoid-responsive element exists between -930 and -910 base pairs relative to the transcription start site. If this element was either deleted or mutated, the negative GC-response was completely lost, which suggests that this sequence is responsible, in part, for the negative regulation of the gene. Electrophoretic mobility shift analysis demonstrated that this palindromic glucocorticoid-responsive element is capable of forming a specific DNA-protein complex with human glucocorticoid receptor. In conclusion, the negative regulation of ALDH3 in rat liver is probably mediated through direct GR binding to its canonical responsive element.


Subject(s)
Aldehyde Dehydrogenase/genetics , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , 5' Untranslated Regions/metabolism , Aldehyde Dehydrogenase/biosynthesis , Animals , Benz(a)Anthracenes/pharmacology , Cells, Cultured , Chloramphenicol O-Acetyltransferase/metabolism , Cycloheximide/pharmacology , Dexamethasone/metabolism , Electrophoresis , Gene Deletion , Genes, Reporter , Glucocorticoids/metabolism , Hormone Antagonists/pharmacology , Humans , Liver/cytology , Liver/drug effects , Male , Mifepristone/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...