Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Pharmacogenetics ; 9(5): 641-50, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10591545

ABSTRACT

Inheritance of the TPMT*2, TPMT*3A and TPMT*3C mutant alleles is associated with deficiency of thiopurine S-methyltransferase (TPMT) activity in humans. However, unlike TPMT*2 and TPMT*3A, the catalytically active protein coded by TPMT*3C does not undergo enhanced proteolysis when heterologously expressed in yeast, making it unclear why this common mutant allele should be associated with inheritance of TPMT-deficiency. To further elucidate the mechanism for TPMT deficiency associated with these alleles, we characterized TPMT proteolysis following heterologous expression of wild-type and mutant proteins in mammalian cells. When expressed in COS-1 cells, proteins encoded by TPMT*2, TPMT*3A, and TPMT*3C cDNAs had significantly reduced steady-state levels and shorter degradation half-lives compared with the wild-type protein. Similarly, in rabbit reticulocyte lysate (RRL), these mutant TPMT proteins were degraded significantly faster than the wild-type protein. Thus, enhanced proteolysis of TPMT*3C protein in mammalian cells is in contrast to its stability in yeast, but consistent with TPMT-deficiency in humans. Proteolysis was ATP-dependent and sensitive to proteasomal inhibitors MG115, MG132 and lactacystin, but not to calpain inhibitor II. We conclude that all of these mutant TPMT proteins undergo enhanced proteolysis in mammalian cells, through an ATP-dependent proteasomal pathway, leading to low or undetectable levels of TPMT protein in humans who inherit these mutant alleles.


Subject(s)
Cysteine Endopeptidases/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , Mutation , Adenosine Triphosphate/metabolism , Alleles , Animals , COS Cells , DNA, Complementary/genetics , Humans , In Vitro Techniques , Kinetics , Methyltransferases/deficiency , Proteasome Endopeptidase Complex , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reticulocytes/metabolism , S-Adenosylmethionine/pharmacology , Transfection
2.
Pharmacogenetics ; 6(4): 279-90, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8873214

ABSTRACT

Thiopurine S-methyltransferase (TPMT) catalyses the S-methylation of thiopurines such as mercaptopurine and thioguanine. TPMT activity exhibits genetic polymorphism, with about 1 in 300 inheriting TPMT-deficiency as an autosomal recessive trait. If treated with standard dosages of thiopurines. TPMT-deficient patients accumulate excessive thioguanine nucleotides (TGN) in hematopoietic tissues, leading to severe hematopoietic toxicity that can be fatal. However, TPMT-deficient patients can be successfully treated with a 10-15-fold lower dosage of these medications. The human gene encoding polymorphic TPMT has been cloned and characterized, and two mutant alleles have recently been isolated from TPMT-deficient and heterozygous patients (TPMT*2, TPMT*3), permitting development of PCR-based methods to identify TPMT-deficient and heterozygous patients prior to therapy. TPMT*3 is the predominant mutant allele in American whites, accounting for about 75% of mutations in this population. Ongoing studies aim to better define the influence of TPMT activity on thiopurine efficacy, to identify additional mutant alleles and determine their frequency in different ethnic groups, to elucidate the mechanism(s) for loss of function of mutant proteins, to identify potential endogenous substrates and to define the molecular mechanisms of TPMT regulation. Together, these advances bold the promise of improving the safety and efficacy of thiopurine therapy.


Subject(s)
Methyltransferases/genetics , Polymorphism, Genetic , Alleles , Humans , Methyltransferases/metabolism , Mutation
3.
Curr Opin Mol Ther ; 2(4): 459-67, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11249778

ABSTRACT

Valspodar (PSC-833) is a derivative of cyclosporin but devoid of the immunosuppressive and nephrotoxic properties seen in cyclosporin A. It exhibited high affinity binding to Mdr1 P-glycoprotein (P-gp) and demonstrated multidrug resistance-reversing activity superior to cyclosporin A and verapamil both in vitro and in vivo. Preclinical and phase I/II clinical data have indicated that plasma levels of PSC-833 with multidrug resistance-reversing activities are achievable. Potent inhibition of intestinal, hepatobiliary and blood-brain barrier P-gp function has been demonstrated. Since valspodar is also a substrate of cytochrome P450 3A (CYP3A), dual interactions of this compound with P-gp and CYP3A are the basis for the pharmacokinetic interactions seen in preclinical and clinical studies. A new formulation of the drug has recently been developed with better oral bioavailability (60%) and less interindividual variability. The toxicity profiles of valspodar are acceptable and dose-limited by transient and reversible cerebellar ataxia. It has shown multidrug resistance-modulating activities towards acute myeloid leukemia, multiple myeloma and ovarian cancer in phase I/II clinical trials. Phase III studies with respect to these three diseases are ongoing.


Subject(s)
Cyclosporins/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Biotechnology , Cyclosporins/adverse effects , Cyclosporins/chemistry , Cyclosporins/metabolism , Drug Resistance, Multiple , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Multiple Myeloma/drug therapy , Ovarian Neoplasms/drug therapy
4.
Clin Biochem ; 26(3): 165-71, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8101143

ABSTRACT

A simple, rapid high-pressure liquid chromatography (HPLC) method was developed for detecting and quantifying benzodiazepines in serum. Seven major benzodiazepines were extracted from spiked serum samples using solid-phase extraction with prazepam as the internal standard. The eluted drugs were then resolved isocratically by HPLC within 11 min using a reversed-phase C8 column with a mobile phase consisting of acetonitrile, methanol, water, and perchloric acid. All drugs gave responses that varied linearly with concentration over the ranges studied. Within-day imprecision (CV) varied from 3.9 to 14.9%, day-to-day CV from 4.8 to 17.0%, absolute recoveries from 67% to 114%, and detection limits from 10 to 110 ng/mL. Tricyclic antidepressants did not interfere, and clinical results were in good agreement with those obtained by a gas chromatographic method. The advantage of this method is that it uses a salt-free isocratic mobile phase that can be easily manipulated to effect difficult benzodiazepine separations.


Subject(s)
Anti-Anxiety Agents/blood , Chromatography, High Pressure Liquid , Humans , Prazepam/blood , Reference Standards
5.
Antivir Chem Chemother ; 10(1): 47-52, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10079879

ABSTRACT

The potency and selectivity index of the AZT-phenyl phosphate derivatives in thymidine kinase (TK)-deficient T cells were substantially enhanced by introducing a single para-bromo substitutent in the phenyl moiety. AZT-5'-(p-bromophenyl methoxyalaninyl phosphate) was 43-fold more potent than AZT-5'-(phenyl methoxyalaninyl phosphate) and was fivefold more potent than AZT in inhibiting human immunodeficiency virus (HIV) replication in TK-deficient CEM cells.


Subject(s)
Anti-HIV Agents/pharmacology , Organophosphorus Compounds/therapeutic use , Zidovudine/analogs & derivatives , Zidovudine/pharmacology , Anti-HIV Agents/chemical synthesis , Dideoxynucleotides , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-1/enzymology , HIV-1/genetics , Humans , Organophosphorus Compounds/pharmacology , Thymine Nucleotides , Virus Replication/drug effects , Virus Replication/genetics , Zidovudine/chemical synthesis , Zidovudine/therapeutic use
6.
J Endourol ; 15(6): 563-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11552776

ABSTRACT

BACKGROUND: The optimal treatment for distal ureteral calculi remains controversial. We present data from our institution to compare the efficacy of extracorporeal shockwave lithotripsy (SWL) and ureteroscopy with different lithotripsy modalities (URSL). METHODS: From January 1994 to September 1997, 954 distal ureteral calculi were treated at our institution using in situ SWL (Siemens Lithostar) in 524 patients and ureteroscopy (Wolf 8.0F instrument and Swiss Lithoclast) in 430 patients. Stone sizes and patient ages were similar in these two groups. RESULTS: In the SWL group, the 3-month stone-free rate was 87%, and the effectiveness quotient (EQ) was 68.7%. In the URSL group, there was a 96% stone-free rate with an EQ of 92.1%. The SWL treatment was more expensive than URSL. CONCLUSION: At our institution, ureteroscopy is more efficacious than SWL for the treatment of distal ureteral calculi. In selected patients who had stones >10 mm with evidence of impaction and severe colic pain, we strongly suggest that URSL is the best choice.


Subject(s)
Lithotripsy , Ureteral Calculi/therapy , Ureteroscopy , Adult , Aged , Aged, 80 and over , Health Care Costs , Humans , Lithotripsy/economics , Lithotripsy/instrumentation , Lithotripsy/methods , Middle Aged , Ureteroscopy/economics
8.
Am J Physiol ; 264(6 Pt 1): C1388-94, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8333493

ABSTRACT

A study was carried out to investigate an ATP-sensitive Ca2+ pool in rat epididymal cells and its role in transepithelial Cl- secretion. In normal buffered solution containing 2.5 mM free Ca2+, ATP triggered single calcium spikes in a dose-dependent fashion. In nominally Ca(2+)-free solution, the peaks of successive Ca2+ spikes diminished after repeated ATP stimulations. Addition of Sr2+ (2.5 mM) to Ca(2+)-free solution after ATP stimulation did not cause changes in fluorescence signals. However, in the presence of Sr2+, ATP gave rise to apparent repetitive Ca2+ spikes of similar magnitudes after repeated stimulations. Increasing the time of exposure in Ca(2+)-free solution containing 50 microM ethylene glycol-bis(beta-amino-ethyl ether)-N,N,N',N'-tetraacetic acid rapidly decreased the intracellular Ca2+ concentration ([Ca2+]i) response to subsequent ATP stimulation. On the other hand, increasing the time of exposure in Sr(2+)-containing solution in Ca(2+)-depleted cells rapidly increased the apparent [Ca2+]i response to subsequent ATP stimulation. These observations suggested the existence of a Ca2+ pool that was rapidly exchanging with the extracellular compartment. Apical application of ATP elicited a transient rise in short-circuit current across the epididymal epithelium in a dose-dependent fashion, and the response was reduced by prior stimulation with thapsigargin. Ca2+ released from a rapidly exchanging ATP-sensitive store might stimulate Cl- secretion in the epididymis, thereby maintaining the electrolyte contents and fluidity of the epididymal microenvironment.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium/metabolism , Epididymis/metabolism , Animals , Cells, Cultured , Electrophysiology , Epididymis/cytology , Epididymis/physiology , Intracellular Membranes/metabolism , Male , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Strontium/pharmacology
9.
Proc Natl Acad Sci U S A ; 94(12): 6444-9, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9177237

ABSTRACT

TPMT is a cytosolic enzyme that catalyzes the S-methylation of aromatic and heterocyclic sulfhydryl compounds, including medications such as mercaptopurine and thioguanine. TPMT activity exhibits autosomal codominant genetic polymorphism, and patients inheriting TPMT deficiency are at high risk of potentially fatal hematopoietic toxicity. The most prevalent mutant alleles associated with TPMT deficiency in humans have been cloned and characterized (TPMT*2 and TPMT*3A), but the mechanisms for loss of catalytic activity have not been elucidated. In the present study, we established that erythrocyte TPMT activity was significantly related to the amount of TPMT protein on Western blots of erythrocytes from patients with TPMT activities of 0.4-23 units/ml pRBC (rs = 0.99; P < 0.001). Similarly, heterologous expression of wild-type (TPMT*1) and mutant (TPMT*2 and TPMT*3A) human cDNAs in yeast and COS-1 cells demonstrated comparable levels of TPMT mRNA but significantly lower TPMT protein with the mutant cDNAs. Rates of protein synthesis were comparable for wild-type and mutant proteins expressed in yeast and with in vitro translation in rabbit reticulocyte lysates. In contrast, pulse-chase experiments revealed significantly shorter degradation half-lives for TPMT*2 and TPMT*3A ( approximately 0.25 hr) compared with wild-type TPMT*1 (18 hr). The degradation of mutant proteins was impaired by ATP depletion and in yeast with mutant proteasomes (pre-1 strain) but unaffected by the lysosomal inhibitor chloroquine. These studies establish enhanced degradation of TPMT proteins encoded by TPMT*2 and TPMT*3A as mechanisms for lower TPMT protein and catalytic activity inherited by the predominant mutant alleles at the human TPMT locus.


Subject(s)
Methyltransferases/genetics , Methyltransferases/metabolism , Polymorphism, Genetic , Alleles , Animals , COS Cells , Cloning, Molecular , Endopeptidases/metabolism , Erythrocytes/enzymology , Humans , Kinetics , Methyltransferases/biosynthesis , Protein Biosynthesis , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Transfection
10.
Pharm Res ; 16(7): 1003-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10450923

ABSTRACT

PURPOSE: To examine the pharmacokinetic features and metabolism of calphostin C, a naturally occurring perylenequinone with potent antileukemic activity. METHODS: HPLC-based quantitative detection methods were used to measure calphostin C levels in lysates of leukemic cells and in plasma of mice treated with calphostin C. The plasma concentration-time data were analyzed using the WinNonlin program. In vitro esterases and a microsome P450 preparation in conjunction with a LC-MS(API-EI) system were used to study the metabolism of calphostin C. RESULTS: An intracellular exposure level (AUC0-6h) of 257 microM x h was achieved after in vitro treatment of NALM-6 cells with calphostin C at a 5 microM final concentration in culture medium. After intraperitoneal (i.p.) injection of a 40 mg/kg nontoxic bolus dose of calphostin C, the estimated Cmax was 2.9 microM, which is higher than the effective in vitro concentration of calphostin C against leukemic cells. Drug absorption after i.p. administration was rapid with an absorption half-life of 24.2 min and the estimated t(max) was 63.0 min. Calphostin C was cleared with an elimination half-life of 91.3 min. An inactive and smaller metabolite (calphostin B) was detected in plasma of calphostin C-treated mice with a t(max) of 41.3 min. Esterase (but not P450) treatment of calphostin C in vitro yielded an inactive metabolite (calphostin B) of the same size and elution profile. CONCLUSIONS: Target plasma calphostin C concentrations of potent antileukemic activity can be reached in mice at nontoxic dose levels. This pilot pharmacokinetic study of calphostin C combined with the availability of the described quantitative HPLC method for its detection in cells and plasma provide the basis for future preclinical evaluation of calphostin C and its potential as an anti-leukemic drug.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacokinetics , Naphthalenes/metabolism , Naphthalenes/pharmacokinetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Chromatography, High Pressure Liquid , Drug Stability , Female , Humans , Mice , Naphthalenes/blood , Naphthalenes/pharmacology , Pilot Projects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Sensitivity and Specificity , Tumor Cells, Cultured
11.
Tech Urol ; 5(4): 191-4, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10591256

ABSTRACT

The aim of this study was to determine the efficacy of intranasal desmopressin in the treatment of nocturnal polyuria in men with benign prostatic hyperplasia (BPH). Twelve men with BPH were treated with intranasal desmopressin at bedtime for nocturnal polyuria. All patients underwent video-urodynamic evaluation. The number of nocturia episodes was the dependent variable. Exclusion criteria included nephrolithiasis, active urinary tact infection, and history of myocardial infarction, congestive heart failure, and angina. Ten of 12 patients improved with the intranasal desmopressin therapy. Nocturia episodes decreased from a median of 3.6 +/- 0.5 episodes/night before treatment to 1.8 +/- 1.1 episodes/night 3 months after therapy (p = .01). The American Urological Association symptom index decreased from 19 +/- 6 before treatment to 12 +/- 6 after therapy (p = .02). Hyponatremia did not occur. We conclude that intranasal desmopressin is a promising therapy for nocturnal polyuria in selected BPH patients.


Subject(s)
Deamino Arginine Vasopressin/administration & dosage , Prostatic Hyperplasia/drug therapy , Renal Agents/administration & dosage , Urination Disorders/drug therapy , Administration, Intranasal , Aged , Drug Administration Schedule , Follow-Up Studies , Humans , Male , Middle Aged , Pilot Projects , Prostatic Hyperplasia/complications , Treatment Outcome , Urination Disorders/etiology , Urodynamics
12.
Bioorg Med Chem Lett ; 8(22): 3121-6, 1998 Nov 17.
Article in English | MEDLINE | ID: mdl-9873688

ABSTRACT

d4T-5'-[p-Bromophenyl methoxyalaninyl phosphate] (d4T-pBPMAP), a novel phenyl phosphate derivative of 2',3'-didehydro-2',3'-dideoxythymidine (d4T) that has an enhanced ability to undergo hydrolysis due to the electron withdrawing properties of its single bromo substituent at the para-position of the phenyl moiety, was found to yield substantially more of the key metabolite alaninyl d4T monophosphate (A-d4T-MP) than the unsubstituted d4T-5'-phenyl methoxyalaninyl phosphate or para-methoxy substituted d4T-5'-phenyl methoxyalaninyl phosphate. d4T-pBPMAP was tested for its anti-HIV-1 activity in peripheral blood mononuclear cells (PBMNC) and thymidine kinase (TK)-deficient CEM T-cells. d4T-pBPMAP was 12.6-fold more potent than the parent compound d4T in inhibiting p24 production (IC50 values: 44 nM vs 556 nM) and 41.3-fold more potent than d4T in inhibiting the reverse transcriptase (RT) activity (IC50 values: 57 nM vs 2355 nM) in HIV-1-infected TK-deficient CEM cells. Similarly, d4T-pBPMAP was more potent than the unsubstituted or para-methoxy substituted phenyl methoxyalaninyl phosphate derivatives of d4T. d4T-pBPMAP did not exhibit any detectable cytotoxicity to PBMNC or CEM cells at concentrations as high as 10,000 nM. Notably, d4T-pBPMAP was capable of inhibiting the replication of a zidovudine (ZDV/AZT)-resistant HIV-1 strain as well as HIV-2 in PBMNC at nanomolar concentrations. To our knowledge, this is the first demonstration that the potency of the d4T-aryl-phosphate derivatives can be substantially enhanced by introducing a single para-bromo substituent in the aryl moiety.


Subject(s)
Anti-HIV Agents/chemical synthesis , Stavudine/analogs & derivatives , Humans , Stavudine/metabolism , Stavudine/pharmacology , Structure-Activity Relationship , Zidovudine/pharmacology
13.
Toxicol Appl Pharmacol ; 140(2): 393-403, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8887457

ABSTRACT

The utilization of precision-cut liver slices in dynamic organ culture as an in vitro model was validated by comparing the induction of the biomarker responses following in vitro (rat liver slice) and in vivo exposure of rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The biomarker responses investigated were cytochrome P450s 1A1 and 1A2 (CYP1A1 and CYP1A2) mRNA, protein, and activities. Precision-cut rat liver slices were incubated in dynamic organ culture for 24 hr with medium containing 0.001-10 nM TCDD or medium without TCDD (control). The resultant mean TCDD concentration in the slices ranged from 19 to 80,925 ppt (wet wt), respectively. A concentration-dependent induction of CYP1A1 mRNA, protein, and activities and a more modest induction of CYP1A2 mRNA was observed in liver slices at all medium concentrations of TCDD. The O-demethylation of 7-methoxyresorufin, a marker for CYP1A2 activity, was induced at TCDD medium levels of 0.01 nM and greater, whereas a detectable increase in CYP1A2 protein occurred only at the higher concentrations. Comparable liver concentrations of TCDD (8-64,698 ppt wet wt) were achieved at 24 hr following a single in vivo exposure of rats to TCDD at doses ranging from 0.002 to 5 microg/kg po. Concentration-effect and dose-response relationships for induction of CYP1A1 and CYP1A2 were similar following in vitro and in vivo exposure to TCDD, although the magnitude of induction was greater for in vivo exposure. The data support the use of liver slices in dynamic organ culture for assessing the relative in vivo potency of a compound to induce CYP1A1 and CYP1A2. Human tissue can also be readily utilized in this in vitro model to predict the biological and toxicological effects of a given in vivo exposure to TCDD.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Liver/drug effects , Liver/enzymology , Polychlorinated Dibenzodioxins/toxicity , Animals , Biomarkers , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A2/drug effects , Enzyme Induction/drug effects , Male , Microtomy , Models, Biological , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
14.
Toxicol Appl Pharmacol ; 123(1): 34-42, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8236259

ABSTRACT

Previous studies have established that TCDF is rapidly metabolized and excreted in rats and that pretreatment of rats with TCDD increases the rate of hepatic metabolism of this compound. The extrahepatic metabolism of TCDF was investigated to assess which enzyme was involved in the metabolism of this compound. Very little metabolism of TCDF was detected in control microsomes (0.3-3.0 pmol/mg/hr), while TCDF metabolism was increased 40- to 200-fold in TCDD-induced rat liver, kidney, and lung microsomes. Since TCDD induces cytochrome P4501A1 and P4501A2 (CYP1A1 and CYP1A2) in the rat liver but only CYP1A1 in kidney and lung, these results suggest that CYP1A1 metabolizes TCDF. To test this hypothesis, TCDF metabolism was investigated in the presence and absence of selective chemical inhibitors and antibodies to CYP1A1 and 1A2. 1-Ethynylpyrene, a suicide inhibitor of CYP1A1 and antibody to rat CYP1A1, produced a dose-dependent inhibition of TCDF metabolism in TCDD-induced rat liver microsomes. Conversely, 2-ethynylnaphthalene, a suicide inhibitor of CYP1A2 and antibody to rat CYP1A2, had no inhibitory effect on the hepatic microsomal metabolism of TCDF. Together, the results strongly indicate that rat CYP1A1 is the primary enzyme responsible for the metabolism of TCDF. 4-Hydroxy-2,3,7,8-TCDF was also identified as the major TCDF metabolite formed by rat CYP1A1. TCDF was also metabolized by human liver microsomes and recombinant yeast microsomes expressing human CYP1A1 and reductase but not by yeast microsomes expressing human CYP1A2 with or without reductase. A similar HPLC profile of TCDF metabolites was observed with microsomes from human liver and yeast expressing human CYP1A1. However, based on ethoxyresorufin-O-deethylase activity, a marker of CYP1A1, the relative rate of TCDF metabolism is about 100-fold greater in TCDD-induced rat liver microsomes than in yeast microsomes expressing human CYP1A1 and reductase. Thus, although TCDF is metabolized by rat and human CYP1A1, the results indicate that there are marked quantitative differences in metabolism which suggest that TCDF will be more persistent in humans.


Subject(s)
Benzofurans/metabolism , Cytochrome P-450 Enzyme System/physiology , Environmental Pollutants/metabolism , Animals , Cytochrome P-450 CYP1A2 , Humans , Liver/metabolism , Male , Oxidoreductases/physiology , Rats , Rats, Sprague-Dawley , Species Specificity
15.
Am J Hum Genet ; 58(4): 694-702, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8644731

ABSTRACT

The autosomal recessive trait of thiopurine S-methytransferase (TPMT) deficiency is associated with severe hematopoietic toxicity when patients are treated with standard doses of mercaptopurine, azathioprine, or thioguanine. To define the molecular mechanism of this genetic polymorphism, we cloned and characterized the cDNA of a TPMT-deficient patient, which revealed a novel mutant allele (TPMT*3) containing two nucleotide transitions (G460-->A and A719-->G) producing amino acid changes at codons 154 (Ala-->Thr) and 240 (Tyr--> Cys), differing from the rare mutant TPMT allele we previously identified (i.e., TPMT*2 with only G238-->C). Site-directed mutagenesis and heterologous expression established that either TPMT*3 mutation alone leads to a reduction in catalytic activity (G460-->A, ninefold reduction; A719-->G, 1.4-fold reduction), while the presence of both mutations leads to complete loss of activity. Using mutation specific PCR-RFLP analysis, the TPMT*3 allele was detected in genomic DNA from approximately 75 percent of unrelated white subjects with heterozygous phenotypes, indicating that TPMT*3 is the most prevalent mutant allele associated with TPMT-deficiency in Caucasians.


Subject(s)
Gene Frequency , Methyltransferases/deficiency , Methyltransferases/genetics , Point Mutation/genetics , White People/genetics , Amino Acid Sequence , Base Sequence , Child, Preschool , Cloning, Molecular , DNA, Complementary/genetics , DNA, Neoplasm/analysis , Enzyme Stability , Erythrocytes/enzymology , Humans , Kinetics , Male , Mercaptopurine/metabolism , Methylation , Methyltransferases/metabolism , Molecular Sequence Data , Polymorphism, Restriction Fragment Length , Precursor Cell Lymphoblastic Leukemia-Lymphoma , RNA, Messenger/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism , Yeasts/genetics
16.
Ann Intern Med ; 126(8): 608-14, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9103127

ABSTRACT

BACKGROUND: Thiopurine S-methyltransferase (TPMT) catalyzes the S-methylation (that is, inactivation) of mercaptopurine, azathioprine, and thioguanine and exhibits genetic polymorphism. About 10% of patients have intermediate TPMT activity because of heterozygosity, and about 1 in 300 inherit TPMT deficiency as an autosomal recessive trait. If they receive standard doses of thiopurine medications (for example, 75 mg/m2 body surface area per day), TPMT-deficient patients accumulate excessive thioguanine nucleotides in hematopoietic tissues, which leads to severe and possibly fatal myelosuppression. OBJECTIVE: To elucidate the genetic basis and develop molecular methods for the diagnosis of TPMT deficiency and heterozygosity. DESIGN: Diagnostic test evaluation. SETTING: Research hospital. PATIENTS: The TPMT phenotype was determined in 282 unrelated white persons, and TPMT genotype was determined in all persons who had intermediate TPMT activity (heterozygotes) and a randomly selected, equal number of persons who had high activity. In addition, genotype was determined in 6 TPMT-deficient patients. MEASUREMENTS: Polymerase chain reaction (PCR) assays were developed to detect the G238C transversion in TPMT*2 and the G460A and A719G transitions in TPMT*3 alleles. Radiochemical assay was used to measure TPMT activity. Mutations of TPMT were identified in genomic DNA, and the concordance of TPMT genotype and phenotype was determined. RESULTS: 21 patients who had a heterozygous phenotype were identified (7.4% of sample [95% CI, 4.7% to 11.2%]). TPMT*3A was the most prevalent mutant allele (18 of 21 mutant alleles in heterozygotes; 85%); TPMT*2 and TPMT*3C were more rare (about 5% each). All 6 patients who had TPMT deficiency had two mutant alleles, 20 of 21 patients (95% [CI, 76% to 99.9%]) who had intermediate TPMT activity had one mutant allele, and 21 of 21 patients (100% [CI, 83% to 100%]) who had high activity had no known TPMT mutation. Detection of TPMT mutations in genomic DNA by PCR coincided perfectly with genotypes detected by complementary DNA sequencing. CONCLUSIONS: The major inactivating mutations at the human TPMT locus have been identified and can be reliably detected by PCR-based methods, which show an excellent concordance between genotype and phenotype. The detection of TPMT mutations provides a molecular diagnostic method for prospectively identifying TPMT-deficient and heterozygous patients.


Subject(s)
Azathioprine/metabolism , Mercaptopurine/metabolism , Methyltransferases/deficiency , Methyltransferases/genetics , Polymerase Chain Reaction/methods , Erythrocytes/enzymology , Genotype , Heterozygote , Humans , Methyltransferases/blood , Mutation , Phenotype , Polymorphism, Genetic
17.
Clin Biochem Rev ; 24(1): 3-12, 2003.
Article in English | MEDLINE | ID: mdl-18568044

ABSTRACT

This mini-review provides a general understanding of electrospray ionisation mass spectrometry (ESI-MS) which has become an increasingly important technique in the clinical laboratory for structural study or quantitative measurement of metabolites in a complex biological sample. The first part of the review explains the electrospray ionisation process, design of mass spectrometers with separation capability, characteristics of the mass spectrum, and practical considerations in quantitative analysis. The second part then focuses on some clinical applications. The capability of ESI-tandem-MS in measuring bio-molecules sharing similar molecular structures makes it particularly useful in screening for inborn errors of amino acid, fatty acid, purine, pyrimidine metabolism and diagnosis of galactosaemia and peroxisomal disorders. Electrospray ionisation is also efficient in generating cluster ions for structural elucidation of macromolecules. This has fostered a new and improved approach (vs electrophoresis) for identification and quantification of haemoglobin variants. With the understanding of glycohaemoglobin structure, an IFCC reference method for glycohaemoglobin assay has been established using ESI-MS. It represents a significant advancement for the standardisation of HbA1c in diabetic monitoring. With its other applications such as in therapeutic drug monitoring, ESI-MS will continue to exert an important influence in the future development and organisation of the clinical laboratory service.

SELECTION OF CITATIONS
SEARCH DETAIL