Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Am Surg ; 89(9): 3959-3961, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37303171

ABSTRACT

Acquired methemoglobinemia is a potentially lethal medical condition caused by exposure to oxidizing xenobiotics, including antibiotics such as dapsone and inhaled anesthetics such as benzocaine. In this case report, we describe two presentations of acquired methemoglobinemia which presented to our surgical intensive care unit within one month. This highlights the potential connection between an emergent surgery or procedure and the development of methemoglobinemia in an environment where it is presumed that this condition would be extremely rare. High clinical suspicion for methemoglobinemia is warranted if the patient develops cyanosis or a decreased oxygen saturation unresponsive to supplemental oxygen when another etiology is not identifiable. If methemoglobinemia is suspected, a direct measurement of blood methemoglobin levels can be obtained to confirm the diagnosis. Prompt treatment with intravenous methylene blue is highly effective.


Subject(s)
Methemoglobinemia , Humans , Methemoglobinemia/chemically induced , Methemoglobinemia/diagnosis , Methylene Blue/therapeutic use , Benzocaine/adverse effects , Cyanosis/complications , Anesthetics, Local/adverse effects , Critical Care
2.
Am Surg ; 88(5): 968-972, 2022 May.
Article in English | MEDLINE | ID: mdl-35187978

ABSTRACT

INTRODUCTION: Opioid use after surgery or trauma has been implicated as a contributing factor to opioid dependence. The Acute Care Surgery (ACS) service at our community-based trauma center instituted an opioid-minimizing, multi-modal pain control (MMPC) protocol. The classes of pain medication included a non-opioid analgesic, a non-steroidal anti-inflammatory drug, a gabapentinoid, a skeletal muscle relaxant, and a topical anesthetic. We hypothesize that the MMPC will result in lower opioid consumption compared with the prior STP as evidenced by lower morphine milligram equivalents (MME) per day. METHODS: All adult patients (≥18 years) admitted to the ACS service from Jan 2014 to Dec 2015 and Jan 2018 to Dec 2019 were screened for inclusion. The standard pain control group (STP) and MMPC groups were defined by the year of admission. The primary outcome is opioid use per day, calculated in MME received. Secondary outcomes of the study include daily pain scores, incidence of opioid-related complications, death, ventilator days, intensive care unit length of stay, and hospital length of stay (HLOS) days. RESULTS: Multi-modal pain control protocol group was older and less injured than STP group. Daily opioid utilization was significantly less in the MMPC group (22.5 MMEs/d vs 60MMEs/d in the STP group, P < .0001). Additionally, daily pain scores were not different between groups. Secondary outcomes did not vary between the two groups. CONCLUSION: This study shows that implementation of a MMPC protocol resulted in lower opioid consumption in injured patients. Pain was equivalently controlled during the MMPC protocol period as demonstrated by similar pain scores.


Subject(s)
Analgesics, Opioid , Opioid-Related Disorders , Adult , Analgesics, Opioid/therapeutic use , Humans , Inpatients , Narcotics/therapeutic use , Pain/drug therapy , Pain/etiology , Pain Management/methods , Pain, Postoperative/drug therapy , Retrospective Studies
3.
Am Surg ; 88(3): 376-379, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34892995

ABSTRACT

INTRODUCTION: The Brain Trauma Foundation advises intracranial pressure monitor placement (ICPM) following traumatic brain injury (TBI) with a Glasgow Coma Scale (GCS) score ≤8 and an abnormal head computed tomographic scan (CT) finding. Prior studies demonstrated that ICPMs could be placed by non-neurosurgeons. We hypothesized that ICPM placement by trauma critical care surgeons (TCCS) would increase appropriate utilization (AU), decrease time to placement (TTP), and have equivalent complications to those placed by neurosurgeons. METHODS: We retrospectively reviewed medical records of adult trauma patients admitted with a TBI in a historical control group (HCG) and practice change group (PCG). Demographics, Injury Severity Score (ISS), outcomes, ICPM placement by provider type, and time to placement were identified. Complications and appropriate utilization were recorded. RESULTS: 70 patients in the HCG and 84 patients in the PCG met criteria for inclusion. Demographics, arrival GCS, ICU GCS, ISS, and admission APACHE II scores were not statistically significant. AU was 7/70 for HCG vs 19/84 in the PCG (P = .04036). Median TTP was 6.5 hours for HCG vs 5.25 for PCG (P = .9308). Interquartile range showed the data clustered around an earlier placement time, 2.3-14.0 hours, in the PCG. Complications between the 2 groups were not statistically significant, 0/7 for HCG vs 5/19 for PCG (P = .2782). DISCUSSION: This study confirms that ICPMs can be safely placed by TCCS. Our results demonstrate that placement of ICPMs by TCCS improves AU and possibly improves TTP.


Subject(s)
Brain Injuries, Traumatic , Intracranial Pressure , Monitoring, Physiologic/instrumentation , Prosthesis Implantation , Surgeons , Traumatology , APACHE , Adult , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnostic imaging , Female , Glasgow Coma Scale , Historically Controlled Study , Humans , Injury Severity Score , Male , Middle Aged , Neurosurgeons , Prosthesis Implantation/adverse effects , Retrospective Studies , Safety , Time-to-Treatment , Tomography, X-Ray Computed , Treatment Outcome
4.
J Biol Chem ; 287(1): 607-618, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22084240

ABSTRACT

Nitrogen monoxide (NO) plays a role in the cytotoxic mechanisms of activated macrophages against tumor cells by inducing iron release. We showed that NO-mediated iron efflux from cells required glutathione (GSH) (Watts, R. N., and Richardson, D. R. (2001) J. Biol. Chem. 276, 4724-4732) and that the GSH-conjugate transporter, multidrug resistance-associated protein 1 (MRP1), mediates this release potentially as a dinitrosyl-dithiol iron complex (DNIC; Watts, R. N., Hawkins, C., Ponka, P., and Richardson, D. R. (2006) Proc. Natl. Acad. Sci. U.S.A. 103, 7670-7675). Recently, glutathione S-transferase P1-1 (GST P1-1) was shown to bind DNICs as dinitrosyl-diglutathionyl iron complexes. Considering this and that GSTs and MRP1 form an integrated detoxification unit with chemotherapeutics, we assessed whether these proteins coordinately regulate storage and transport of DNICs as long lived NO intermediates. Cells transfected with GSTP1 (but not GSTA1 or GSTM1) significantly decreased NO-mediated 59Fe release from cells. This NO-mediated 59Fe efflux and the effect of GST P1-1 on preventing this were observed with NO-generating agents and also in cells transfected with inducible nitric oxide synthase. Notably, 59Fe accumulated in cells within GST P1-1-containing fractions, indicating an alteration in intracellular 59Fe distribution. Furthermore, electron paramagnetic resonance studies showed that MCF7-VP cells transfected with GSTP1 contain significantly greater levels of a unique DNIC signal. These investigations indicate that GST P1-1 acts to sequester NO as DNICs, reducing their transport out of the cell by MRP1. Cell proliferation studies demonstrated the importance of the combined effect of GST P1-1 and MRP1 in protecting cells from the cytotoxic effects of NO. Thus, the DNIC storage function of GST P1-1 and ability of MRP1 to efflux DNICs are vital in protection against NO cytotoxicity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Glutathione S-Transferase pi/metabolism , Iron/metabolism , Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Biological Transport/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glutathione S-Transferase pi/deficiency , Glutathione S-Transferase pi/genetics , Intracellular Space/drug effects , Intracellular Space/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type II/metabolism , Propionates/pharmacology , Quinolines/pharmacology , S-Nitrosoglutathione/metabolism , Transfection
5.
Chem Biol Interact ; 194(2-3): 113-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21925487

ABSTRACT

4-Hydroxy-2-nonenal (HNE) is one of the most reactive products of lipid peroxidation and has both cytotoxic and genotoxic effects in cells. Several enzymatic pathways have been reported to detoxify HNE, including conjugation by glutathione-S-transferases (GSTs). Removal of the resulting HNE-glutathione conjugate (HNE-SG) by an efflux transporter may be required for complete detoxification. We investigated the effect of expression of GSTM1 and/or the ABC efflux transporter protein, multidrug-resistance protein-1 (MRP1), on HNE-induced cellular toxicity. Stably transfected MCF7 cell lines were used to examine the effect of GSTM1 and/or MRP1 expression on HNE-induced cytotoxicity, GSH depletion, and HNE-protein adduct formation. Co-expression in the MCF7 cell line of GSTM1 with MRP1 resulted in a 2.3-fold sensitization to HNE cytotoxicity (0.44-fold IC(50) value relative to control) rather than the expected protection. Expression of either GSTM1 or MRP1 alone also resulted in slight sensitization to HNE cytotoxicity (0.79-fold and 0.71-fold decreases in IC(50) values, respectively). Co-expression of GSTM1 and MRP1 strongly enhanced the formation of HNE-protein adducts relative to the non-expressing control cell line, whereas expression of either MRP1 alone or GSTM1 alone yielded similarly low levels of HNE-protein adducts to that of the control cell line. Glutathione (GSH) levels were reduced by 10-20% in either the control cell line or the MCF7/GSTM1 cell line with the same HNE exposure for 60min. However, HNE induced >80% depletion of GSH in cells expressing MRP1 alone. Co-expression of both MRP1 and GSTM1 caused slightly greater GSH depletion, consistent with the greater protein adduct formation and cytotoxicity in this cell line. Since expression of GSTM1 or MRP1 alone did not strongly sensitize cells to HNE, or result in greater HNE-protein adducts than in the control cell line, these results indicate that MRP1 and GSTM1 collaborate to enhance HNE-protein adduct formation and HNE cytotoxicity, facilitated by GSH depletion mediated by both MRP1 and GSTM1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aldehydes/pharmacology , Glutathione Transferase/metabolism , Glutathione/metabolism , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Humans , Inhibitory Concentration 50
6.
Biochemistry ; 50(36): 7765-73, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21827153

ABSTRACT

Naturally occurring nitroalkene fatty acids (NAs) derived from oleic (NO(2)-OA) and linoleic (NO(2)-LA) acids mediate a variety of cellular responses. We examined the signaling pathways involved in NA activation of Nrf2/ARE-dependent versus PPARγ/PPRE-dependent transcription in human MCF7 breast cancer cells. Additionally, we compared the relative potencies of NO(2)-OA and NO(2)-LA in activating these two transcriptional programs. Here it is demonstrated that, in addition to the direct adduct formation of NA with the Nrf2 inhibitory protein, Keap1, shown by others, NA activation of Nrf2/ARE-mediated transcription results from increased nuclear Nrf2 levels and depends upon activation of the PI3K/AKT and PKC, but not ERK and JNK MAPK, signaling pathways. Examination of the relationship between NA stimulation of the Nrf2/ARE versus PPARγ/PPRE transcriptional programs revealed concentration-dependent activation of distinct signaling pathways that were readily distinguished by selective attenuation of Nrf2/ARE-dependent, but not PPARγ-dependent, transcription by inhibitors of PI3K and PKC. Moreover, measurable, statistically significant activation of PPARγ/PPRE-dependent transcription occurred at nanomolar concentrations of NAs-the 12-NO(2) isomer of NO(2)-LA showing the most potent activity-whereas significant activation of Nrf2/ARE-dependent transcription occurred at much higher NA concentrations (≥3 µM) with the NO(2)-OA isomers the most potent. These findings have implications for the physiological roles of NAs, suggesting that, at concentrations likely to be encountered in vivo, their direct activation of PPARγ transcription will dominate over their electrophilic activation of Nrf2 antioxidant/protective responses.


Subject(s)
Linoleic Acids/pharmacology , NF-E2-Related Factor 2/metabolism , Oleic Acids/pharmacology , PPAR gamma/metabolism , Response Elements , Signal Transduction , Transcription, Genetic , Alkenes/chemistry , Cell Line, Tumor , Humans , Linoleic Acids/chemistry , MAP Kinase Kinase 4/metabolism , NF-E2-Related Factor 2/genetics , Oleic Acids/chemistry , PPAR gamma/genetics
7.
J Med Chem ; 52(15): 4631-9, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19719236

ABSTRACT

Nitroalkene fatty acids are potent endogenous ligand activators of PPARgamma-dependent transcription. Previous studies with the naturally occurring regioisomers of nitrolinoleic acid revealed that the isomers are not equivalent with respect to PPARgamma activation. To gain further insight into the structure-activity relationships between nitroalkenes and PPARgamma, we examined additional naturally occurring nitroalkenes derived from oleic acid, 9-nitrooleic acid (E-9-NO2-18:1 [1]) and 10-nitrooleic acid (E-10-NO2-18:1 [2]), and several synthetic nitrated enoic fatty acids of variable carbon chain length, double bonds, and nitration site. At submicromolar concentrations, E-12-NO2 derivatives were considerably more potent than isomers nitrated at carbons 5, 6, 9, 10, and 13, and chain length (16 versus 18) or number of double bonds (1 versus 2) was of little consequence for PPARgamma activation. Interestingly, at higher concentrations (>2 microM) the nitrated enoic fatty acids (E-9-NO2-18:1 [1], E-9-NO2-16:1 [3], E-10-NO2-18:1 [2], and E-12-NO2-18:1 [7]) deviated significantly from the saturable pattern of PPARgamma activation observed for nitrated 1,4-dienoic fatty acids (E-9-NO2-18:2, E-10-NO2-18:2, E-12-NO2-18:2, and E-13-NO2-18:2).


Subject(s)
Alkenes/chemical synthesis , Fatty Acids/chemical synthesis , PPAR gamma/drug effects , Alkenes/metabolism , Alkenes/pharmacology , Cell Line, Tumor , Fatty Acids/metabolism , Fatty Acids/pharmacology , Humans , PPAR gamma/metabolism , Structure-Activity Relationship
8.
Biochemistry ; 48(19): 4159-69, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19358561

ABSTRACT

The naturally occurring nitroalkenes, nitrolinoleic (NO(2)-LA) and nitrooleic (NO(2)-OA) acids, are among the most potent endogenous ligand activators of PPARgamma-dependent transcription. In order to understand mechanisms that regulate cellular response to these nitroalkenes, we previously demonstrated that glutathione conjugation of NO(2)-LA and MRP1-mediated efflux of the conjugates were associated with significant attenuation of PPARgamma activation by this nitroalkene [(2006) Biochemistry 45, 7889-7896]. Here we show that NO(2)-OA activation of PPARgamma is similarly affected by nonenzymatic conjugation and MRP1-mediated efflux. Moreover, the roles of glutathione S-transferases (GSTs) in the glutathione conjugation and bioactivities of NO(2)-LA and NO(2)-OA were investigated. While none of the GST isozymes tested (GSTA1-1, A4-4, M1a-1a, and P1a-1a) enhanced the rate of glutathione conjugation, expression of GSTA1-1, M1a-1a, or P1a-1a in MCF7 cells significantly reduced the magnitude of PPARgamma-dependent reporter gene transcription in response to NO(2)-LA and NO(2)-OA treatment, with GSTP1a-1a expression mediating the most potent inhibition of PPARgamma. Although these GSTs failed to catalyze nitroalkene conjugation with glutathione, the nitroalkenes were found to associate avidly with all four GST isozymes as indicated by their ability to inhibit GST activity with K(i)'s in the nanomolar range. Treatment of purified GSTP1a-1a with excess NO(2)-LA and NO(2)-OA resulted in the formation of covalent adducts between GSTP1a monomers and nitroalkenes, although separate experiments indicated that such covalent bond formation was not necessary for avid GST-nitroalkene interactions. These results suggest that GSTs can inhibit the activation of transcription by nitroalkenes via noncatalytic sequestration of these ligands, and their glutathione conjugates, away from their nuclear target, PPARgamma.


Subject(s)
Alkenes/pharmacology , Fatty Acids/pharmacology , Glutathione Transferase/antagonists & inhibitors , Nitro Compounds/pharmacology , PPAR gamma/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/pharmacology , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Spectrophotometry, Ultraviolet
9.
Chem Biol Interact ; 179(2-3): 240-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19330882

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). We have used V79MZ cells stably transfected with either human or rat cytochrome P4501A1 (CYP1A1), alone or in combination with human GSTP1 (hGSTP1), to examine the dynamics of activation versus detoxification of benzo[a]pyrene (B[a]P), dibenzo[a,l]pyrene (DB[a,l]P), and their dihydrodiol metabolites. The cytotoxicity of B[a]P or DB[a,l]P was 9-11-fold greater in cells expressing human, as compared to rat CYP1A1, despite similar enzymatic activities. Co-expression of the hGSTP1 with the hCYP1A1 conferred 16-fold resistance to B[a]P cytotoxicity, compared to only 2.5-fold resistance when hGSTP1 was co-expressed with rat CYP1A1. The lower B[a]P cytotoxicity in the cells expressing rat CYP1A1, and weaker protection by hGSTP1 co-expression in these cells, were attributable to the much lower fraction of B[a]P metabolism via formation of the 7,8-dihydrodiol intermediate by the rat CYP1A1 compared to hCYP1A1. Resistance to the DB[a,l]P cytotoxicity conferred by hGSTP1 expression was also greater in cells co-expressing hCYP1A1 (7-fold) as compared to cells co-expressing rCYP1A1 (<2-fold). Resistance to B[a]P conferred by hGSTP1 was closely correlated with the activity level in two clonal transfectant lines with a 3-fold difference in hGSTP1-1 specific activity. Depletion of GSH to 20% of control levels via pretreatment with the de novo GSH biosynthesis inhibitor buthionine sulfoximine reduced the protection against B[a]P cytotoxicity by hGSTP1 from 16-fold to 5-fold, indicating that catalysis of conjugation with GSH, rather than binding or other effects, is responsible for the resistance. The cytotoxicity of the dihydrodiol intermediates of B[a]P or DB[a,l]P was much greater, and similar in cell lines expressing either human or rat CYP1A1. Again, however, the protection conferred by hGSTP1 co-expression was 2-5-fold greater in cells with hCYP1A1 than with rCYP1A1 expression. These results indicate that GST expression can effectively limit cytotoxicity following activation of B[a]P by human or rat CYP1A1, but is less effective as a defense against exposure of cells to the intermediate metabolite B[a]P-7,8-dihydrodiol.


Subject(s)
Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dihydroxydihydrobenzopyrenes/toxicity , Glutathione S-Transferase pi/metabolism , Naphthalenes/toxicity , Transgenes/genetics , Animals , Benzo(a)pyrene/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cytochrome P-450 CYP1A1/biosynthesis , Dihydroxydihydrobenzopyrenes/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Naphthalenes/metabolism , Rats
10.
Biochemistry ; 48(2): 492-8, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19105608

ABSTRACT

Previous studies demonstrated that the naturally occurring electrophile and PPARgamma ligand, nitrolinoleic acid (NO(2)-LA), exists as a mixture of four regioisomers [Alexander, R. L., et al. (2006) Biochemistry 45, 7889-7896]. We hypothesized that these alternative isomers have distinct bioactivities; therefore, to determine if the regioisomers are quantitatively or qualitatively different with respect to PPARgamma activation, NO(2)-LA was separated into three fractions which were identified by NMR (13-NO(2)-LA, 12-NO(2)-LA, and a mixture of 9- and 10-NO(2)-LA) and characterized for PPARgamma interactions. A competition radioligand binding assay showed that all three NO(2)-LA fractions had similar binding affinities for PPARgamma (IC(50) = 0.41-0.60 microM) that were comparable to that of the pharmaceutical ligand, rosiglitazone (IC(50) = 0.25 microM). However, when PPARgamma-dependent transcription activation was examined, there were significant differences observed among the NO(2)-LA fractions. Each isomer behaved as a partial agonist in this reporter gene assay; however, the 12-NO(2) derivative was the most potent with respect to maximum activation of PPARgamma and an EC(50) of 0.045 microM (compare with the rosiglitazone EC(50) of 0.067 microM), while the 13-NO(2) and 9- and 10-NO(2) derivatives were considerably less effective with EC(50) values of 0.41-0.62 microM. We conclude that the regioisomers of NO(2)-LA are not functionally equivalent. The 12-NO(2) derivative appears to be the most potent in PPARgamma-dependent transcription activation, whereas the weaker PPARgamma agonists, 13-NO(2) and 9- and 10-NO(2), may be relatively more important in signaling via other, PPARgamma-independent pathways in which this family of nitrolipid electrophiles is implicated.


Subject(s)
Linoleic Acids/pharmacology , Nitro Compounds/pharmacology , PPAR gamma/metabolism , Binding, Competitive , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Clone Cells , Dose-Response Relationship, Drug , Female , Genes, Reporter , Humans , Inhibitory Concentration 50 , Linoleic Acids/chemistry , Luciferases/metabolism , Nitro Compounds/chemistry , Nuclear Magnetic Resonance, Biomolecular , PPAR gamma/agonists , PPAR gamma/genetics , Radioligand Assay , Rosiglitazone , Stereoisomerism , Thiazolidinediones/metabolism , Transcriptional Activation/drug effects , Transduction, Genetic
11.
Toxicol Lett ; 183(1-3): 99-104, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18992797

ABSTRACT

The environmental carcinogen 5-methylchrysene (5MC) can be activated to mutagenic metabolites by several isozymes of cytochrome P-450 (CYP). The resulting reactive diol-epoxides can be detoxified via conjugation by glutathione S-transferases (GST). We investigated whether expression of human glutathione S-transferase P1 (hGSTP1) would differentially protect cells against the cytotoxicity or mutagenicity of 5MC or its 1,2-dihydrodiol intermediate (5MC-1,2-diol) in V79MZ cells with activation via stably transfected human CYP1B1 (hCYP1B1) as compared to activation by human CYP1A1 (hCYP1A1). The parent compound 5MC was only 2-fold more cytotoxic in the CYP-expressing cell lines than in the V79MZ parental cell line, while 5MC-1,2-dihydrodiol was more than 30-fold more cytotoxic in CYP-transfected cells compared to V79MZ cells. Cells co-expressing either hCYP1B1 or hCYP1A1 together with hGSTP1 were 2-fold less sensitive to 5MC or 5MC-1,2-diol cytotoxicity than their CYP-only parent lines. The 5MC was highly mutagenic with similar potency in both hCYP-transfected cell lines, while 5MC-1,2-diol was 2-fold more mutagenic in hCYP1B1-transfected cells as compared to hCYP1A1 cells. Coexpression of hGSTP1 with either hCYP reduced 5MC or 5MC-1,2-diol mutagenicity by 1.4-4.5-fold compared to the corresponding hCYP-only expressing cell lines. The greater protection against mutagenicity of 5MC is in contrast to our previous studies in which we found greater protection by hGSTP1 against cytotoxicity than mutagenicity of benzo[a]pyrene in cells co-expressing hCYP1A1. Protection against mutagenicity by hGSTP1 was greater with activation of either compound by hCYP1B1 than with hCYP1A1 activation. These studies show that the relative efficacy of protection by hGSTP1 against mutagenicity of 5MC or 5MC-1,2-diol is in part determined by the specific CYP pathway that catalyzes activation to the toxic or mutagenic metabolites.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Chrysenes/toxicity , Cytochrome P-450 CYP1A1/metabolism , Glutathione S-Transferase pi/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Carcinogens/chemistry , Carcinogens/toxicity , Cell Line , Cell Survival/drug effects , Chrysenes/chemistry , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1 , Dose-Response Relationship, Drug , Glutathione S-Transferase pi/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Inhibitory Concentration 50 , Mutagenicity Tests/methods , Transfection
12.
Mol Cancer Ther ; 7(10): 3247-55, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18852128

ABSTRACT

Cells expressing elevated levels of allelic variants of human glutathione S-transferase P1 (GSTP1) and/or efflux transporters, MRP1 or MRP2, were used to evaluate the role of GSTP1-1 in cisplatin resistance. These studies revealed that GSTP1-1 confers low-level resistance (1.4- to 1.7-fold) to cisplatin-induced cytotoxicity in MCF7 cells. However, expression of MRP1 (MCF7 cells) or MRP2 (HepG2 cells) failed to augment or potentiate GSTP1-1-mediated resistance in either cell line. To understand the mechanism by which variants of GSTP1-1 confer resistance to cisplatin, their relative abilities to catalyze conjugation of cisplatin with glutathione were examined. Enzymes encoded by all three alleles tested, GSTP1a (I(104)A(113)), GSTP1b (V(104)A(113)), and GSTP1c (V(104)V(113)), increased the formation rate of the mono-platinum/glutathione derivative of cisplatin with relative catalytic activities of 1.0 (GSTP1a-1a variant) and 1.8 to 1.9 (GSTP1b-1b and GSTP1c-1c variants). Although these data are consistent with the idea that very low level resistance to cisplatin may be conferred by GSTP1-1-mediated cisplatin/glutathione conjugation, two observations indicate that such catalysis plays a minor role in the protection from cisplatin toxicity. First, the rates of GSTP1-1-mediated conjugation are extremely slow (1.7-2.6 h(-1) at 25 degrees C). Second, despite an 80% to 90% increase in catalysis of cisplatin conjugation by GSTP1b-1b or GSTP1c-1c over GSTP1a-1a, we observed no discernable differences in relative resistances conferred by these alternative variants when expressed in MCF7 cells. We conclude that high-level cisplatin resistance attributed to GSTP1-1 in other studies is not likely due to catalysis of cisplatin conjugation but rather must be explained by other mechanisms, which may include GSTP1-mediated modulation of signaling pathways.


Subject(s)
Cisplatin/metabolism , Glutathione S-Transferase pi/metabolism , Alleles , Catalysis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Glutathione/chemistry , Glutathione/metabolism , Humans , Inactivation, Metabolic , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mass Spectrometry , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Transduction, Genetic
13.
J Trauma ; 64(6): 1638-50, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18545134

ABSTRACT

The American College of Surgeons Committee on Trauma's Advanced Trauma Life Support Course is currently taught in 50 countries. The 8th edition has been revised following broad input by the International ATLS subcommittee. Graded levels of evidence were used to evaluate and approve changes to the course content. New materials related to principles of disaster management have been added. ATLS is a common language teaching one safe way of initial trauma assessment and management.


Subject(s)
Curriculum/standards , Education, Medical, Continuing , Life Support Care/standards , Traumatology/education , Wounds and Injuries/therapy , Clinical Competence , Curriculum/trends , Emergency Medicine/education , Emergency Treatment/standards , Emergency Treatment/trends , Female , Forecasting , Humans , Life Support Care/trends , Male , Resuscitation/education , Sensitivity and Specificity , Traumatology/trends , United States
14.
Carcinogenesis ; 29(4): 807-15, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18204073

ABSTRACT

A major component of the anticarcinogenic activity of the dietary chemopreventive agent sulforaphane (SFN) is attributed to its ability to induce expression of phase II detoxification genes containing the antioxidant response element (ARE) within their promoters. Because SFN is a reactive electrophile--readily forming conjugates with glutathione (GSH)--we asked whether expression of glutathione S-transferase (GST) P1-1 and the GSH conjugate efflux pump, multidrug resistance or resistance-associated protein (MRP) 1, would significantly modify the cellular response to SFN exposure. This was investigated using GST- and MRP1-poor parental MCF7 cells and transgenic derivatives expressing GSTP1-1 and/or MRP1. Compared with parental cells, expression of GSTP1-1 alone enhanced the rate of intracellular accumulation of SFN and its glutathione conjugate, SFN-SG--an effect that was associated with increased ARE-containing reporter gene induction. Expression of MRP1 greatly reduced SFN/SFN-SG accumulation and resulted in significant attenuation of SFN-mediated induction of ARE-containing reporter and endogenous gene expression. Coexpression of GSTP1-1 with MRP1 further reduced the level of induction. Depletion of GSH prior to SFN treatment or the substitution of tert-butylhydroquinone for SFN abolished the effects of MRP1/GSTP1-1 on ARE-containing gene induction-indicating that these effects are GSH dependent. Lastly, analysis of NF-E2-related factor 2 (Nrf2)--a transcription factor operating via binding to the ARE--showed that the increased levels of Nrf2 following SFN treatment were considerably less sustained in MRP1-expressing, especially those coexpressing GSTP1-1, than in MRP1-poor cells. These results suggest that the regulating effects of MRP1 and GSTP1-1 expression on SFN-dependent induction of phase II genes are ultimately mediated by altering nuclear Nrf2 levels.


Subject(s)
Anticarcinogenic Agents/pharmacology , Glutathione S-Transferase pi/genetics , Multidrug Resistance-Associated Proteins/genetics , Thiocyanates/pharmacology , Biological Transport/drug effects , Breast Neoplasms , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Glutathione/metabolism , Glutathione S-Transferase pi/drug effects , Humans , Isothiocyanates , Multidrug Resistance-Associated Proteins/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Sulfoxides , Transcriptional Activation
15.
Mutat Res ; 624(1-2): 80-7, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17509623

ABSTRACT

We have used V79MZ hamster lung fibroblasts stably transfected with human cytochrome P450-1A1 (hCYP1A1; cell line designated V79MZh1A1) or P450-1B1 (hCYP1B1; cell line designated V79MZh1B1) alone, or in combination with human glutathione-S-transferase (GST) alpha-1 (hGSTA1), in order to examine GST protection against cytotoxicity and mutagenicity of dibenzo[a,l]pyrene (DBP) and the intermediate dihydrodiol metabolite (+/-)-DBP-11,12-dihydrodiol (DBPD). At comparable expression levels of hCYP1A1 and hCYP1B1, both DBP and DBPD were more cytotoxic in V79MZ1A1 (IC(50)=2.7 and 0.7nM, respectively) than in V79MZh1B1 (IC(50)=6.0 and 4.8nM, respectively). In contrast, both DBP and DBPD were two- to four-fold more mutagenic in V79MZh1B1 than in V79MZ1A1. Co-expression of hGSTA1 with hCYP1A1 decreased DBP cytotoxicity two-fold compared to V79MZh1A1 with hCYP1A1 alone, and provided a small, yet still statistically significant, 1.3-fold protection against DBPD. Protection against mutagenicity of these compounds was comparable to that for cytotoxicity in cells expressing hCYP1A1. In V79MZh1B1 cells, co-expression of hGSTA1 conferred up to five-fold protection against DBP cytotoxicity, and up to nine-fold protection against the (+/-)-DBP-dihydrodiol cytotoxicity relative to the cells expressing hCYP1B1 alone. Co-expression of hGSTA1 also reduced mutagenicity of DBP or its dihydrodiol to a lesser extent (1.3-1.8-fold) than the protection against cytotoxicity in cells expressing hCYP1B1. These findings demonstrate that the protective efficacy of hGSTA1 against DBP and DBPD toxicity is variable, depending on the compound or metabolite present, the specific cytochrome P450 isozyme expressed, and the specific cellular damage endpoint examined.


Subject(s)
Benzopyrenes/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 Enzyme System/genetics , Dihydroxydihydrobenzopyrenes/toxicity , Glutathione Transferase/genetics , Mutagens/toxicity , Animals , Aryl Hydrocarbon Hydroxylases , Benzopyrenes/metabolism , Cell Line , Cell Survival/drug effects , Cricetinae , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme System/metabolism , Dihydroxydihydrobenzopyrenes/metabolism , Glutathione Transferase/metabolism , Humans , Mutagens/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
16.
Toxicol Sci ; 99(1): 51-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17525473

ABSTRACT

Transgenic cell lines were constructed to study the dynamics of competition between activation versus detoxification of benzo[a]pyrene (B[a]P) or B[a]P-7,8-dihydrodiol metabolites. Stably transfected V79MZ cells expressing human cytochrome P4501A1 (hCYP1A1) alone or in combination with human glutathione-S-transferase M1 (hGSTM1) were used to determine how effectively this GST isozyme protects against cytotoxic, genotoxic, and mutagenic effects of B[a]P or the enantiomeric dihydrodiol metabolites (+)-benzo[a]pyrene-7,8-dihydrodiol ((+)-B[a]P-7,8-diol) and (-)-benzo[a]pyrene-7,8-dihydrodiol ((-)-B[a]P-7,8-diol). Expression of hGSTM1 in the presence of hCYP1A1 conferred significant 8.5-fold protection against B[a]P-induced cytotoxicity, but protection against cytotoxicity of either B[a]P-7,8-diol enantiomer was not significant. Mutagenicity of B[a]P at the hprt locus was dose and time dependent in cells that expressed hCYP1A1. Mutagenicity of B[a]P was reduced by 21-32% and mutagenicity induced by the B[a]P-7,8-diols was reduced 20-58% in cells further modified to coexpress hGSTM1-1 compared to cells expressing hCYP1A1 alone. Expression of hGSTM1-1 reduced adducts in total cellular macromolecules by twofold, in good correlation with the reduction in B[a]P mutagenicity. These results indicate that while hGSTM1-1 effectively protects against hCYP1A1-mediated cytotoxicity of B[a]P, a significant fraction of the mutagenicity that results from activation of B[a]P and its 7,8-dihydrodiol metabolites by hCYP1A1 is derived from B[a]P metabolites that are not detoxified by hGSTM1.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/metabolism , DNA Adducts/metabolism , Dihydroxydihydrobenzopyrenes/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Glutathione Transferase/metabolism , Mutagens/toxicity , Animals , Benzo(a)pyrene/pharmacokinetics , Cell Line , Cricetinae , Cricetulus , Cytochrome P-450 CYP1A1/genetics , DNA/drug effects , DNA/genetics , Dihydroxydihydrobenzopyrenes/pharmacokinetics , Fibroblasts/drug effects , Fibroblasts/enzymology , Glutathione Transferase/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/drug effects , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Inactivation, Metabolic , Mutagenicity Tests , Mutagens/pharmacokinetics , Mutation , Stereoisomerism , Transfection
17.
Carcinogenesis ; 28(1): 207-14, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16885195

ABSTRACT

Transgenic cell lines were constructed to study dynamic competition between activation versus detoxification of benzo[a]pyrene (B[a]P) and its metabolites. Transfected V79MZ cells expressing human cytochrome P4501A1 (hCYP1A1) alone, or expressing hCYP1A1 in combination with human glutathione S-transferase P1 (hGSTP1), were used to determine how effectively GST protects against macromolecular damage or mutagenicity of B[a]P or its enantiomeric dihydrodiol metabolites (+)-benzo[a]pyrene-7,8-dihydrodiol [(+)B[a]P-7,8-diol] and (-)-benzo[a]pyrene-7,8-dihydrodiol [(-)-B[a]P-7,8-diol]. Mutagenicity of B[a]P at the hprt locus was dose- and time-dependent in cells that expressed hCYP1A1. Mutagenicity was reduced in cells further modified to co-express hGSTP1. Dose-response and time-course studies indicated that mutagenicity was reduced up to 3-fold by hGSTP1 expression, compared with cells expressing hCYP1A1 alone. Mutagenicity induced by the B[a]P 7,8-dihydrodiols was also dose-dependent, and was reduced 2- to 5-fold by hGSTP1. Expression of hGSTP1 reduced B[a]P adducts in total cellular macromolecules by 3.8-fold, which correlated with the reduction in B[a]P mutagenicity and with reduction in the formation of the proximate metabolite B[a]P 7,8-dihydrodiols from B[a]P. However, measurement of total B[a]P metabolites bound to DNA isolated from cells incubated with [3H]-B[a]P revealed only 12, 33 and 24% reduction at 12, 24 and 48 h, respectively, by GSTP1 expression. Nevertheless, (32)P-post-labeling analysis demonstrated nearly total prevention of the known B[a]P-DNA adduct, N2-guanine-benzo[a]pyrene-7,8-diol-9,10-epoxide (BPDE), in cells co-expressing hGSTP1. This adduct, thought to be the most mutagenic of the stable B[a]P adducts, accounts for 15% or less of the total DNA adducts observed. These results indicate that the reduction in hCYP1A1-mediated B[a]P mutagenesis by hGSTP1 is probably largely due to prevention of the N2-guanine-BPDE adduct. However, the significant fraction (30-40%) of this mutagenesis and the majority of the total DNA binding that are not prevented together suggest formation by hCYP1A1 of a subset of mutagenic metabolites of B[a]P that are not effectively detoxified by hGSTP1.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/metabolism , DNA Adducts , DNA Damage , Glutathione S-Transferase pi/metabolism , Mutagenesis , Mutagens/toxicity , Alkylation , Animals , Cells, Cultured , Cricetinae , Cricetulus , Cytochrome P-450 CYP1A1/genetics , Glutathione S-Transferase pi/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/physiology , Transfection
18.
Biochemistry ; 45(25): 7889-96, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16784241

ABSTRACT

Recent data has shown that nitrolinoleic acid (LNO(2)), an electrophilic derivative of linoleic acid, has several important bioactivities including antiinflammatory, antiplatelet, vasorelaxation, and-as a novel potent ligand of PPARgamma-transcription regulating activities. Moreover, LNO(2) is formed in abundance in vivo at levels sufficient to mediate these bioactivities. In order to investigate the role of glutathione conjugation and MRP1-mediated efflux in the regulation of PPARgamma-dependent LNO(2) signaling, regioisomers of LNO(2) were synthesized and characterized. Analysis by 1D and 2D (1)H and (13)C NMR revealed that the LNO(2) preparation consisted of four, rather than two, nitrated regioisomers in approximately equal abundance. At physiologic pH and intracellular glutathione levels, LNO(2) was rapidly and quantitatively converted to glutathione conjugates (LNO(2)-SG) via Michael addition. MRP1 mediated efficient ATP-dependent transport of LNO(2)-SG. Using a PPRE-containing reporter gene transiently transfected into MRP-poor MCF7/WT cells, we verified that the LNO(2) mixture was a potent activator of PPARgamma-dependent transcription. However, expression of MRP1 in the stably transduced MCF7 derivative, MCF7/MRP1-10, resulted in strong inhibition of LNO(2)-induced transcription activation. Taken together, these results suggest that glutathione conjugation and MRP1-mediated conjugate transport can attenuate LNO(2) bioactivity and thereby play important roles in the regulation of cellular signaling by LNO(2).


Subject(s)
Linoleic Acids/pharmacology , Multidrug Resistance-Associated Proteins/physiology , Nitro Compounds/pharmacology , PPAR gamma/physiology , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Cell Membrane/physiology , Glutathione/metabolism , Humans , Linoleic Acids/metabolism , Nitro Compounds/metabolism , Nuclear Magnetic Resonance, Biomolecular , Tumor Cells, Cultured
19.
Mol Pharmacol ; 69(4): 1499-505, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16434618

ABSTRACT

Based upon several previous reports, no consistent relationship between multidrug resistance protein 1 (MRP1, ABCC1) expression and cellular sensitivity to mitoxantrone (MX) toxicity can be ascertained; thus, the role of MRP1 in MX resistance remains controversial. The present study, using paired parental, MRP1-poor, and transduced MRP1-overexpressing MCF7 cells, unequivocally demonstrates that MRP1 confers resistance to MX cytotoxicity and that resistance is associated with reduced cellular accumulation of MX. This MRP1-associated reduced accumulation of MX was partially reversed by treatment of cells with 50 microM MK571 [3-[[3-[2-(7-chloroquinolin-2-yl)vinyl]phenyl]-(2-dimethylcarbamoylethylsulfanyl)methylsulfanyl] propionic acid]-an MRP inhibitor that increased MX accumulation in MRP1-expressing MCF7 cells but had no effect on MRP-poor MCF7 cells. Moreover, in vitro experiments using inside-out membrane vesicles show that MRP1 supports ATP-dependent, osmotically sensitive uptake of MX. Unlike ABCG2 (breast cancer resistance protein, mitoxantrone-resistant protein), MRP1-mediated MX transport is dependent upon the presence of glutathione or its S-methyl analog. In addition, MX stimulates transport of [3H]glutathione. Together, these data are consistent with the interpretation that MX efflux by MRP1 involves cotransport of MX and glutathione. The results suggest that MRP1-like the alternative MX transporters ABCG2 and ABCB1 (MDR1, P-glycoprotein)-can significantly influence tumor cell sensitivity to and pharmacological disposition of MX.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Glutathione/metabolism , Mitoxantrone/pharmacology , Multidrug Resistance-Associated Proteins/physiology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Mitoxantrone/pharmacokinetics , Propionates/pharmacology , Quinolines/pharmacology
20.
Biochim Biophys Acta ; 1736(3): 228-36, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16154383

ABSTRACT

MDA-MB-231, MCF7, and SKOV3 cancer cells, but not HEK-293 cells, expressed mRNA for the leukocyte G protein-coupled 5-oxo-eicosatetraenoate (ETE) OXE receptor. 5-Oxo-ETE, 5-oxo-15-OH-ETE, and 5-HETE stimulated the cancer cell lines but not HEK-293 cells to mount pertussis toxin-sensitive proliferation responses. Their potencies in eliciting this response were similar to their known potencies in activating leukocytes and OXE receptor-transfected cells. However, high concentrations of 5-oxo-ETE and 5-oxo-15-OH-ETE, but not 5-HETE, arrested growth and caused apoptosis in all four cell lines; these responses were pertussis toxin-resistant. The same high concentrations of the oxo-ETEs but again not 5-HETE also activated peroxisome proliferator-activated receptor (PPAR)-gamma. Pharmacological studies indicated that this activation did not mediate their effects on proliferation. These results are the first to implicate the OXE receptor in malignant cell growth and to show that 5-oxo-ETEs activate cell death programs as well as PPARgamma independently of this receptor.


Subject(s)
Arachidonic Acids/pharmacology , Cell Proliferation/drug effects , Receptors, Eicosanoid/physiology , Anilides/pharmacology , Apoptosis/drug effects , Arachidonic Acids/metabolism , Binding Sites/genetics , Caspase 3 , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression/genetics , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Hydroxyeicosatetraenoic Acids/pharmacology , Mitosis/drug effects , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Pertussis Toxin/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Protein Binding , Receptors, Eicosanoid/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...