70 results on '"Finch RA"'
Search Results
2. Treatment of Goodpastureʼs Syndrome With Immunosuppression and Plasmapheresis
- Author
-
Wilson Cb, Finch Ra, Edwin A. Rutsky, and McGowan E
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Cyclophosphamide ,Anti-Glomerular Basement Membrane Disease ,medicine.medical_treatment ,Kidney Glomerulus ,Renal function ,urologic and male genital diseases ,Gastroenterology ,Antibodies ,Basement Membrane ,Prednisone ,Internal medicine ,medicine ,Goodpasture's syndrome ,Humans ,biology ,business.industry ,Immunosuppression ,Plasmapheresis ,General Medicine ,biology.protein ,Antibody ,business ,medicine.drug - Abstract
A 14-year-old boy with Goodpasture's syndrome induced by anti-glomerular-basement-membrane (gmb) antibody exhibited declining renal function, in association with a progressive increase in the level of serum anti-GBM antibody. Treatment with prednisone, cyclophosphamide, and plasmapheresis was associated with rapid disappearance of the serum anti-GMB antibody and temporary stabilization of renal function.
- Published
- 1979
3. Ionized calcium levels in umbilical cord blood of women with preeclampsia and normotensive pregnancies.
- Author
-
Yusuf K, Kamaluddeen M, Hasan SU, Al-Awad E, Finch RA, and Akierman AR
- Published
- 2012
4. Hypernatremia during lithium and ticarcillin therapy
- Author
-
Finch Ra
- Subjects
medicine.medical_specialty ,Lithium (medication) ,Penicillins ,Lithium ,Sepsis ,chemistry.chemical_compound ,Polyuria ,Lithium Carbonate ,medicine ,Humans ,Ticarcillin ,Intensive care medicine ,Aged ,Hypernatremia ,business.industry ,Lithium carbonate ,General Medicine ,medicine.disease ,Leukemia ,chemistry ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
A patient being treated for leukemia received lithium carbonate and ticarcillin for sepsis, and polyuria and severe hypernatremia developed. Although useful in neutropenic patients, the simultaneous use of these drugs may result in life-threatening hypernatremia.
- Published
- 1981
5. Anti-tumor activity of camptothecin analog conjugate of a RSPO4-based peptibody targeting LGR4/5/6 in preclinical models of colorectal cancer.
- Author
-
Toh Y, Wu L, Tu J, Liang Z, Aldana AM, Li L, Wen JJ, Pan S, Julie RH, Hensel ME, Hodo CL, Finch RA, Carmon KS, and Liu QJ
- Abstract
Antibody-drug conjugates (ADCs) have emerged as a major modality of targeted cancer therapy, yet no ADC has been approved for colorectal cancer (CRC). LGR4/5/6 (leucine-rich repeat containing, G protein-coupled receptor 4, 5, 6) are three related receptors that are expressed at high levels together or alternately in nearly all cases of CRC. ADCs targeting LGR5 have been shown to have robust anti-tumor potency, but not all CRC cells express LGR5 and LGR5-positive tumor cells may lose LGR5 expression due to cancer cell plasticity. R-spondin 4 (RSPO4) is a natural protein ligand of LGR4/5/6 with high affinity for all three receptors. We fused a mutant form of RSPO4 that retains high affinity binding to LGR4/5/6 to IgG1 Fc to create a peptibody designated R462. Conjugation of R462 with a camptothecin analog (CPT2) at eight drugs per peptibody led to the synthesis of R462-CPT2 that showed highly potent cytotoxic activity in vitro in CRC cell lines expressing any of LG4/5/6. In cell line xenograft and PDX models of CRC, R462-CPT2 demonstrated robust anti-tumor effect. Importantly, R462-CPT2 showed no major adverse effect at therapeutically effective dose levels. These results strongly support the use of RSPO ligand drug-conjugates that target LGR4/5/6 simultaneously for the treatment of CRC.
- Published
- 2024
- Full Text
- View/download PDF
6. pH-dependent general base catalyzed activation rather than isocyanate liberation may explain the superior anticancer efficacy of laromustine compared to related 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine prodrugs.
- Author
-
Penketh PG, Finch RA, Sauro R, Baumann RP, Ratner ES, and Shyam K
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Catalysis, Cell Line, Tumor, DNA chemistry, DNA metabolism, Drug Design, Female, Half-Life, Humans, Hydrazines pharmacokinetics, Hydrazines pharmacology, Hydrazines therapeutic use, Hydrogen-Ion Concentration, Leukemia drug therapy, Leukemia pathology, Mice, Prodrugs pharmacology, Prodrugs therapeutic use, Protein Carbamylation, Sulfonamides pharmacokinetics, Sulfonamides pharmacology, Sulfonamides therapeutic use, Transplantation, Homologous, Antineoplastic Agents chemistry, Hydrazines chemistry, Isocyanates metabolism, Prodrugs chemistry, Sulfonamides chemistry
- Abstract
Laromustine (also known as cloretazine, onrigin, VNP40101M, 101M) is a prodrug of 90CE, a short-lived chloroethylating agent with anticancer activity. The short half-life of 90CE necessitates the use of latentiated prodrug forms for in vivo treatments. Alkylaminocarbonyl-based prodrugs such as laromustine exhibit significantly superior in vivo activity in several murine tumor models compared to analogs utilizing acyl, and alkoxycarbonyl latentiating groups. The alkylaminocarbonyl prodrugs possess two exclusive characteristics: (i) They are primarily unmasked by spontaneous base catalyzed elimination; and (ii) they liberate a reactive carbamoylating species. Previous speculations as to the therapeutic superiority of laromustine have focused upon the inhibition of enzymes by carbamoylation. We have investigated the therapeutic interactions of analogs with segregated chloroethylating and carbamoylating activities (singly and in combination) in the in vivo murine L1210 leukemia model. The combined treatment with chloroethylating and carbamoylating prodrugs failed to result in any synergism and produced a reduction in the therapeutic efficacy compared to the chloroethylating prodrug alone. Evidence supporting an alternative explanation for the superior tumor selectivity of laromustine is presented that is centered upon the high pH sensitivity of its base catalyzed activation, and the more alkaline intracellular pH values commonly found within tumor cells., (© 2017 John Wiley & Sons A/S.)
- Published
- 2018
- Full Text
- View/download PDF
7. Preclinical Evaluation of the Short-Term Toxicity of 4-(N)-Docosahexaenoyl 2´, 2´- Difluorodeoxycytidine (DHA-dFdC).
- Author
-
Valdes S, Naguib YW, Finch RA, Baze WB, Jolly CA, and Cui Z
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Deoxycytidine pharmacology, Drug Compounding, Female, Humans, Maximum Tolerated Dose, Mice, Inbred DBA, Gemcitabine, Antineoplastic Agents toxicity, Deoxycytidine analogs & derivatives, Deoxycytidine toxicity, Leukemia L1210 drug therapy
- Abstract
Purpose: This study was designed to test the short-term toxicity of DHA-dFdC in a mouse model and its efficacy in a mouse model of leukemia at or below its repeat-dose maximum tolerated dose (RD-MTD)., Method: A repeat-dose dose-ranging toxicity study was designed to determine the tolerability of DHA-dFdC when administered to DBA/2 mice by intravenous (i.v.) injection on a repeat-dose schedule (i.e. injections on days 0, 3, 7, 10, and 13). In order to determine the effect of a lethal dose of DHA-dFdC, mice were injected i.v. with three doses of DHA-dFdC at 100 mg/kg on days 0, 3, and 5 (i.e. a lethal-RD). The body weight of mice was recorded two or three times a week. At the end of the study, major organs (i.e. heart, liver, spleen, kidneys, lung, and pancreas) of mice that received the lethal-RD or RD-MTD were weighed, and blood samples were collected for analyses. Finally, DHA-dFdC was i.v. injected into DBA/2 mice with syngeneic L1210 mouse leukemia cells to evaluate its efficacy at or below RD-MTD., Results: The RD-MTD of DHA-dFdC is 50 mg/kg. At 100 mg/kg, a lethal-RD, DHA-dFdC decreases the weights of mouse spleen and liver and significantly affected certain blood parameters (i.e. white blood cells, lymphocytes, eosinophils, and neutrophil segmented). At or below its RD-MTD, DHA-dFdC significantly prolonged the survival of L1210 leukemia-bearing mice., Conclusion: DHA-dFdC has dose-dependent toxicity, affecting mainly spleen at a lethal-RD. At or below its RD-MTD, DHA-dFdC is effective against leukemia in a mouse model.
- Published
- 2017
- Full Text
- View/download PDF
8. Antitumor sulfonylhydrazines: design, structure-activity relationships, resistance mechanisms, and strategies for improving therapeutic utility.
- Author
-
Shyam K, Penketh PG, Baumann RP, Finch RA, Zhu R, Zhu YL, and Sartorelli AC
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Hypoxia, Chemotherapy, Cancer, Regional Perfusion, Combined Modality Therapy, DNA Modification Methylases metabolism, DNA Repair Enzymes metabolism, Drug Design, Humans, Hydrazines pharmacology, Hydrazines therapeutic use, Precision Medicine, Structure-Activity Relationship, Sulfones pharmacology, Sulfones therapeutic use, Tumor Suppressor Proteins metabolism, Antineoplastic Agents chemistry, Drug Resistance, Neoplasm, Hydrazines chemistry, Sulfones chemistry
- Abstract
1,2-Bis(sulfonyl)-1-alkylhydrazines (BSHs) were conceived as more specific DNA guanine O-6 methylating and chloroethylating agents lacking many of the undesirable toxicophores contained in antitumor nitrosoureas. O(6)-Alkylguanine-DNA alkyltransferase (MGMT) is the sole repair protein for O(6)-alkylguanine lesions in DNA and has been reported to be absent in 5-20% of most tumor types. Many BSHs exhibit highly selective cytotoxicity toward cells deficient in MGMT activity. The development of clinically useful MGMT assays should permit the identification of tumors with this vulnerability and allow for the preselection of patient subpopulations with a high probability of responding. The BSH system is highly versatile, permitting the synthesis of many prodrug types with the ability to incorporate an additional level of tumor-targeting due to preferential activation by tumor cells. Furthermore, it may be possible to expand the spectrum of activity of these agents to include tumors with MGMT activity by combining them with tumor-targeted MGMT inhibitors.
- Published
- 2015
- Full Text
- View/download PDF
9. Triplex-forming oligonucleotides targeting c-MYC potentiate the anti-tumor activity of gemcitabine in a mouse model of human cancer.
- Author
-
Boulware SB, Christensen LA, Thames H, Coghlan L, Vasquez KM, and Finch RA
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols, Chromatin Immunoprecipitation, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Deoxycytidine pharmacology, Female, Humans, Mice, Mice, Nude, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Gemcitabine, Antimetabolites, Antineoplastic pharmacology, Colonic Neoplasms prevention & control, DNA, Neoplasm genetics, Deoxycytidine analogs & derivatives, Drug Synergism, Oligonucleotides pharmacology, Proto-Oncogene Proteins c-myc antagonists & inhibitors
- Abstract
Antimetabolite chemotherapy remains an essential cancer treatment modality, but often produces only marginal benefit due to the lack of tumor specificity, the development of drug resistance, and the refractoriness of slowly proliferating cells in solid tumors. Here, we report a novel strategy to circumvent the proliferation-dependence of traditional antimetabolite-based therapies. Triplex-forming oligonucleotides (TFOs) were used to target site-specific DNA damage to the human c-MYC oncogene, thereby inducing replication-independent, unscheduled DNA repair synthesis (UDS) preferentially in the TFO-targeted region. The TFO-directed UDS facilitated incorporation of the antimetabolite, gemcitabine (GEM), into the damaged oncogene, thereby potentiating the anti-tumor activity of GEM. Mice bearing COLO 320DM human colon cancer xenografts (containing amplified c-MYC) were treated with a TFO targeted to c-MYC in combination with GEM. Tumor growth inhibition produced by the combination was significantly greater than with either TFO or GEM alone. Specific TFO binding to the genomic c-MYC gene was demonstrated, and TFO-induced DNA damage was confirmed by NBS1 accumulation, supporting a mechanism of enhanced efficacy of GEM via TFO-targeted DNA damage-induced UDS. Thus, coupling antimetabolite chemotherapeutics with a strategy that facilitates selective targeting of cells containing amplification of cancer-relevant genes can improve their activity against solid tumors, while possibly minimizing host toxicity., (© 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
10. Targeting oncogenes to improve breast cancer chemotherapy.
- Author
-
Christensen LA, Finch RA, Booker AJ, and Vasquez KM
- Subjects
- Base Sequence, Cell Adhesion physiology, Cell Growth Processes drug effects, Cell Growth Processes genetics, Cell Line, Tumor, DNA Damage, Deoxycytidine pharmacology, Drug Synergism, Gene Expression drug effects, Humans, Molecular Sequence Data, Oligonucleotides genetics, Transcription, Genetic drug effects, Transcription, Genetic genetics, Gemcitabine, Adenocarcinoma drug therapy, Adenocarcinoma genetics, Antimetabolites, Antineoplastic pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Deoxycytidine analogs & derivatives, Genes, myc drug effects, Oligonucleotides pharmacology
- Abstract
Despite recent advances in treatment, breast cancer remains a serious health threat for women. Traditional chemotherapies are limited by a lack of specificity for tumor cells and the cell cycle dependence of many chemotherapeutic agents. Here we report a novel strategy to help overcome these limitations. Using triplex-forming oligonucleotides (TFOs) to direct DNA damage site-specifically to oncogenes overexpressed in human breast cancer cells, we show that the effectiveness of the anticancer nucleoside analogue gemcitabine can be improved significantly. TFOs targeted to the promoter region of c-myc directly inhibited gene expression by approximately 40%. When used in combination, specific TFOs increased the incorporation of gemcitabine at the targeted site approximately 4-fold, presumably due to induction of replication-independent DNA synthesis. Cells treated with TFOs and gemcitabine in combination showed a reduction in both cell survival and capacity for anchorage-independent growth (approximately 19% of untreated cells). This combination affected the tumorigenic potential of these cancer cells to a significantly greater extent than either treatment alone. This novel strategy may be used to increase the range of effectiveness of antitumor nucleosides in any tumor which overexpresses a targetable oncogene. Multifaceted chemotherapeutic approaches such as this, coupled with triplex-directed gene targeting, may lead to more than incremental improvements in nonsurgical treatment of breast tumors.
- Published
- 2006
- Full Text
- View/download PDF
11. Age-related differences in vincristine toxicity and biodistribution in wild-type and transporter-deficient mice.
- Author
-
Muramatsu T, Johnson DR, Finch RA, Johnson LK, Leffert JJ, Lin ZP, Pizzorno G, and Sartorelli AC
- Subjects
- ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters physiology, Age Factors, Animals, Female, Genotype, Male, Mice, Reverse Transcriptase Polymerase Chain Reaction, Antineoplastic Agents, Phytogenic pharmacokinetics, Antineoplastic Agents, Phytogenic toxicity, Genes, MDR genetics, Vincristine pharmacokinetics, Vincristine toxicity
- Abstract
The impact of mouse multidrug resistance genes mdrla/b and mrpl on age-related differences in the toxicity and biodistribution of vincristine (VCR) was evaluated in wild-type, mrpl(-/-), mdrla/b(-/-), and combined mdrla/b(-/-), mrpl(-/-) weanling and adult mice given a single IP dose of VCR ranging from 0.0625 to 6 mg/kg. Weanling mice of all four genotypes were more sensitive than adult animals as determined by survival rate, average time of death, and pathologic findings. Wild-type animals were the least sensitive and combined mdrla/b(-/-), mrpl(-/-) mice the most sensitive to VCR toxicity. Mdrla/b(-/-) and mrpl(-/-) genotypes exhibited intermediate sensitivities, with mdrla/b(-/-) mice being more sensitive than mrpl(-/-) animals to the vinca alkaloid. Administration of [3H]VCR to wild-type and mdrla/b(-/-), mrpl(-/-) animals revealed relatively greater accumulation of radioactive VCR equivalents in weanlings over adults in several tissues, with weanling mdrla/b(-/-), mrpl(-/-) lung and heart exhibiting the greatest enhanced accumulation of 26- and 15-fold over adults, respectively. A similar cardiopulmonary differential accumulation of VCR was not observed in wild-type weanlings to adults. Semiquantitative RT-PCR expression analyses of ABC transporter genes in weanling and adult tissues of wild-type and combined mdrla/b(-/-), mrpl(-/-) mice did not reveal major age-related differences in these ABC transporters that would explain the relatively greater toxicity observed in weanling mice. However, the greater cardiopulmonary accumulation of VCR equivalents seen in the combined mdrla/b(-/-), mrpl(-/-) weanlings over that of adults underscores the potential for unique organ and age-related toxicities of this agent in the setting of transporter deficiency.
- Published
- 2004
- Full Text
- View/download PDF
12. Wnk1 kinase deficiency lowers blood pressure in mice: a gene-trap screen to identify potential targets for therapeutic intervention.
- Author
-
Zambrowicz BP, Abuin A, Ramirez-Solis R, Richter LJ, Piggott J, BeltrandelRio H, Buxton EC, Edwards J, Finch RA, Friddle CJ, Gupta A, Hansen G, Hu Y, Huang W, Jaing C, Key BW Jr, Kipp P, Kohlhauff B, Ma ZQ, Markesich D, Payne R, Potter DG, Qian N, Shaw J, Schrick J, Shi ZZ, Sparks MJ, Van Sligtenhorst I, Vogel P, Walke W, Xu N, Zhu Q, Person C, and Sands AT
- Subjects
- Animals, Base Sequence, Blood Pressure genetics, DNA, Complementary genetics, Gene Library, Genetic Techniques, Heterozygote, Humans, Hypertension therapy, Intracellular Signaling Peptides and Proteins, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Minor Histocompatibility Antigens, Molecular Sequence Data, Mutagenesis, Insertional methods, Phenotype, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases physiology, Sequence Tagged Sites, WNK Lysine-Deficient Protein Kinase 1, Blood Pressure physiology, Protein Serine-Threonine Kinases deficiency
- Abstract
The availability of both the mouse and human genome sequences allows for the systematic discovery of human gene function through the use of the mouse as a model system. To accelerate the genetic determination of gene function, we have developed a sequence-tagged gene-trap library of >270,000 mouse embryonic stem cell clones representing mutations in approximately 60% of mammalian genes. Through the generation and phenotypic analysis of knockout mice from this resource, we are undertaking a functional screen to identify genes regulating physiological parameters such as blood pressure. As part of this screen, mice deficient for the Wnk1 kinase gene were generated and analyzed. Genetic studies in humans have shown that large intronic deletions in WNK1 lead to its overexpression and are responsible for pseudohypoaldosteronism type II, an autosomal dominant disorder characterized by hypertension, increased renal salt reabsorption, and impaired K+ and H+ excretion. Consistent with the human genetic studies, Wnk1 heterozygous mice displayed a significant decrease in blood pressure. Mice homozygous for the Wnk1 mutation died during embryonic development before day 13 of gestation. These results demonstrate that Wnk1 is a regulator of blood pressure critical for development and illustrate the utility of a functional screen driven by a sequence-based mutagenesis approach.
- Published
- 2003
- Full Text
- View/download PDF
13. High-throughput mouse knockouts provide a functional analysis of the genome.
- Author
-
Friddle CJ, Abuin A, Ramirez-Solis R, Richter LJ, Buxton EC, Edwards J, Finch RA, Gupta A, Hansen G, Holt KH, Hu Y, Huang W, Jaing C, Key BW Jr, Kipp P, Kohlhauff B, Ma ZQ, Markesich D, Newhouse M, Perry T, Platt KA, Potter DG, Qian N, Shaw J, Schrick J, Shi ZZ, Sparks MJ, Tran D, Wann ER, Walke W, Wallace JD, Xu N, Zhu Q, Person C, Sands AT, and Zambrowicz BP
- Subjects
- Animals, Base Sequence, DNA genetics, Gene Library, Genomics methods, Mice, Knockout, Molecular Sequence Data, Sequence Tagged Sites, Genome, Mice genetics
- Published
- 2003
- Full Text
- View/download PDF
14. Comparative study of the importance of multidrug resistance-associated protein 1 and P-glycoprotein to drug sensitivity in immortalized mouse embryonic fibroblasts.
- Author
-
Lin ZP, Johnson DR, Finch RA, Belinsky MG, Kruh GD, and Sartorelli AC
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Animals, Anticarcinogenic Agents pharmacology, Antineoplastic Agents pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Biological Transport, Blotting, Western, Cell Survival, Cells, Cultured, Dose-Response Relationship, Drug, Etoposide pharmacology, Fluoresceins metabolism, Fluoresceins pharmacology, Inhibitory Concentration 50, Mice, Mice, Knockout, Precipitin Tests, RNA metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Transfection, Tumor Cells, Cultured, Vincristine pharmacology, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Chemokines, CC physiology, Fibroblasts metabolism
- Abstract
Multidrug resistance-associated protein 1 and P-glycoprotein are major ATP-binding cassette transporters that function as efflux pumps and confer resistance to a variety of structurally unrelated anticancer agents. To evaluate the comparative importance of these transporters with respect to anticancer agents, we established and characterized SV40-immortalized [mrp1(-/-)] (KO), [mdr1a/1b(-/-)] (DKO), and combined [mrp1 (-/-), mdr1a/1b(-/-)] (TKO) deficient fibroblast lines derived from primary embryonic fibroblasts of knockout mice. Western blot analyses demonstrated that KO and DKO fibroblasts exhibited similar levels of P-glycoprotein and mrp1, respectively, to that of wild-type (WT) fibroblasts. In addition, semiquantitative reverse transcription-PCR measurements of other multidrug resistance-associated protein (mrp) family members demonstrated that TKO fibroblasts displayed expression profiles of mrps 2-7 comparable to that of WT fibroblasts. These results indicate that loss of mrp1, P-glycoprotein, or both transporters does not cause overt compensatory changes in the expression of the other determined transporters. Using cell viability and calcein accumulation assays, we demonstrated that KO and DKO fibroblasts exhibited a low to moderate increase in sensitivity to vincristine and etoposide and in calcein accumulation compared to WT fibroblasts, whereas TKO fibroblasts displayed a markedly enhanced sensitivity to these agents and further elevated calcein accumulation. Furthermore, verapamil, an inhibitor of both mrp1 and P-glycoprotein, significantly sensitized WT fibroblasts to both vincristine and etoposide while having no effect on the sensitivity of TKO cells to these agents. Collectively, these findings indicate that mrp1 and P-glycoprotein are major determinants of drug sensitivity in immortalized mouse embryonic fibroblasts. They also suggest the existence of a compensatory mechanism by which the loss of one transporter can be functionally offset by the other in the transport of common drug substrates.
- Published
- 2002
15. mdmx is a negative regulator of p53 activity in vivo.
- Author
-
Finch RA, Donoviel DB, Potter D, Shi M, Fan A, Freed DD, Wang CY, Zambrowicz BP, Ramirez-Solis R, Sands AT, and Zhang N
- Subjects
- Animals, Apoptosis physiology, Cell Division physiology, Cells, Cultured, Embryo, Mammalian, Fibroblasts cytology, Fibroblasts physiology, Mice, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Nuclear Proteins, Proto-Oncogene Proteins physiology, Tumor Suppressor Protein p53 physiology
- Abstract
Regulation of p53 protein activity is required for normal embryogenesis, tumor suppression, and cellular response to DNA damage. Here we report that loss of mdmx, a p53-binding protein, results in midgestational embryo lethality, a phenotype that is completely rescued by the absence of p53. Mice homozygous for both mdmx and p53 null mutations are viable and appear developmentally normal. Fibroblasts derived from embryos with reduced mdmx expression demonstrate a decreased growth rate and increased UV-induced apoptosis compared with wild-type cells and contain elevated levels of p53 and several p53 target proteins including the proapoptotic bax protein. These observations demonstrate that mdmx functions as a critical negative regulator of p53 in vivo.
- Published
- 2002
16. Human XPA and RPA DNA repair proteins participate in specific recognition of triplex-induced helical distortions.
- Author
-
Vasquez KM, Christensen J, Li L, Finch RA, and Glazer PM
- Subjects
- Base Sequence, Binding Sites, DNA Primers pharmacology, DNA Repair, Dose-Response Relationship, Drug, Enzyme-Linked Immunosorbent Assay, Ficusin chemistry, Humans, Introns, Molecular Sequence Data, Mutagenesis, Site-Directed, Protein Binding, Recombination, Genetic, Replication Protein A, Ultraviolet Rays, Xeroderma Pigmentosum Group A Protein, DNA metabolism, DNA Damage, DNA-Binding Proteins physiology
- Abstract
Nucleotide excision repair (NER) plays a central role in maintaining genomic integrity by detecting and repairing a wide variety of DNA lesions. Xeroderma pigmentosum complementation group A protein (XPA) is an essential component of the repair machinery, and it is thought to be involved in the initial step as a DNA damage recognition and/or confirmation factor. Human replication protein A (RPA) and XPA have been reported to interact to form a DNA damage recognition complex with greater specificity for damaged DNA than XPA alone. The mechanism by which these two proteins recognize such a wide array of structures resulting from different types of DNA damage is not known. One possibility is that they recognize a common feature of the lesions, such as distortions of the helical backbone. We have tested this idea by determining whether human XPA and RPA proteins can recognize the helical distortions induced by a DNA triple helix, a noncanonical DNA structure that has been shown to induce DNA repair, mutagenesis, and recombination. We measured binding of XPA and RPA, together or separately, to substrates containing triplexes with three, two, or no strands covalently linked by psoralen conjugation and photoaddition. We found that RPA alone recognizes all covalent triplex structures, but also forms multivalent nonspecific DNA aggregates at higher concentrations. XPA by itself does not recognize the substrates, but it binds them in the presence of RPA. Addition of XPA decreases the nonspecific DNA aggregate formation. These results support the hypothesis that the NER machinery is targeted to helical distortions and demonstrate that RPA can recognize damaged DNA even without XPA.
- Published
- 2002
- Full Text
- View/download PDF
17. Transport of fluorescein in MDCKII-MRP1 transfected cells and mrp1-knockout mice.
- Author
-
Sun H, Johnson DR, Finch RA, Sartorelli AC, Miller DW, and Elmquist WF
- Subjects
- ATP-Binding Cassette Transporters genetics, Animals, Biological Transport drug effects, Blood-Brain Barrier, Dogs, Kinetics, Mice, Mice, Knockout, Multidrug Resistance-Associated Proteins, Probenecid pharmacology, Propionates pharmacology, Quinolines pharmacology, Recombinant Proteins metabolism, Substrate Specificity, Tissue Distribution, Transfection, ATP-Binding Cassette Transporters metabolism, Base Pair Mismatch, Brain metabolism, Fluorescein pharmacokinetics
- Abstract
The multidrug resistant-associated protein 1 (MRP1) is a membrane-bound transport protein that is involved in the efflux of organic anions and has been implicated in multidrug resistance in cancer. MRP1 has also been reported to be ubiquitously expressed in normal tissues, including the brain. The presence of functional organic anion transporters in the blood-brain and blood-CSF barriers that influence the distribution of various compounds to the brain has long been known. The purpose of this study was to examine the role of MRP1 in the brain distribution of a model organic anion, fluorescein. The substrate specificity of MRP1 for fluorescein was initially determined by examining the accumulation of fluorescein in MDCKII MRP1-transfected cells. The distribution of fluorescein in the brain was then examined in wild-type and mrp1 gene knockout mice. The results show that in MDCKII MRP1-transfected cells, the accumulation of fluorescein was significantly lower (about 40% lower) than that in wild-type MDCKII cells. MRP1 inhibitors such as probenecid, MK-571, and LY402913 enhanced fluorescein accumulation in MDCKII MRP1-transfected cells to a greater extent than in wild-type MDCKII cells. In an in vivo study, after intravenous injection of fluorescein, the fluorescein brain-to-plasma concentration ratio in mrp1 knockout mice was not significantly different than that in wild-type mice. However, when probenecid was co-administered with fluorescein in wild-type mice, the fluorescein brain-to-plasma ratio was significantly increased (1.5-fold). These findings suggest that fluorescein is a substrate for MRP1. Furthermore, the in vivo study also suggests that MRP1 has a limited role in the transport and distribution of fluorescein in the brain. Therefore, other organic anion transport proteins, including the various isoforms of the MRP family, may be responsible for the accumulation and transport of organic anions in the brain., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
18. Optimization of an exogenous metabolic activation system for FETAX. II. Preliminary evaluation.
- Author
-
Fort DJ, Rogers RL, Paul RR, Stover EL, and Finch RA
- Subjects
- Abnormalities, Drug-Induced etiology, Animals, Carbon Tetrachloride toxicity, Chloroform toxicity, Coculture Techniques, Isoniazid, Microsomes, Liver enzymology, Microsomes, Liver metabolism, Parathion toxicity, Phenacetin toxicity, Phenobarbital, Rats, Urethane toxicity, beta-Naphthoflavone, Biotransformation, Embryo, Mammalian drug effects, Embryo, Nonmammalian, Toxicity Tests, Xenopus laevis embryology
- Abstract
The developmental toxicities of five test compounds including carbon tetrachloride, urethane, phenacetin, parathion, and chloroform, were evaluated using Frog Embryo Teratogenesis Assay--Xenopus (FETAX), with minor modification. Post-isolation mixtures of differently-induced rat liver microsomes (phenobarbital- (PB), beta-naphthoflavone- (beta-NF), and isoniazid- (INH)-induced preparations) were co-cultured directly with X. laevis embryos. Results from these studies suggest that the Aroclor 1254-induced MAS could effectively be replaced by a mixed lot of PB-, beta-NF-, and INH-induced rat liver microsomes. Each of the test materials were found to be developmentally toxic when bioactivated by the mixed MAS.
- Published
- 2001
- Full Text
- View/download PDF
19. Optimization of an exogenous metabolic activation system for FETAX. I. Post-isolation rat liver microsome mixtures.
- Author
-
Fort DJ, Rogers RL, Stover EL, and Finch RA
- Subjects
- 2-Acetylaminofluorene toxicity, Abnormalities, Drug-Induced etiology, Animals, Chlorodiphenyl (54% Chlorine), Coumarins toxicity, Cyclophosphamide toxicity, Isoniazid, Microsomes, Liver enzymology, Microsomes, Liver metabolism, Phenobarbital, Rats, Trichloroethylene toxicity, beta-Naphthoflavone, Biotransformation, Embryo, Mammalian drug effects, Embryo, Nonmammalian, Toxicity Tests, Xenopus laevis embryology
- Abstract
The developmental toxicity of cyclophosphamide, coumarin, 2-acetyl-aminofluorine (2-AAF), and trichloroethylene (TCE) was assessed with Frog Embryo Teratogenesis Assay: Xenopus (FETAX). Late Xenopus laevis blastulae were exposed to each test material for 96-h in two separate static-renewal tests with and without the presence of five differently induced exogenous metabolic activation systems (MAS). The MAS consisted of Aroclor 1254- (Aroclor 1254 MAS), isoniazid- (INH MAS), phenobarbital- (PB MAS), or beta-naphthoflavone- (beta-NF MAS), or a post-isolation mixture (mixed MAS) of INH-, PB-, and beta-NF-induced rat liver microsomes. Addition of the Aroclor 1254 MAS bioactivated cyclophosphamide, coumarin, 2-AAF, but not TCE. Addition of the PB MAS bioactivated cyclophosphamide, weakly bioactivated coumarin and 2-AAF, but had no effect on TCE developmental toxicity. The beta-NF MAS bioactivated coumarin and 2-AAF, weakly bioactivated cyclophosphamide, but did not alter the developmental toxicity of TCE. Addition of the INH-induced MAS only bioactivated TCE, whereas the post-isolation mixed MAS bioactivated each test material. Based on LC50 and EC50 (malformation) values, embryo growth, and types and severity of induced malformations, each test material was developmentally toxic. Use of post-microsome isolation mixtures from differentially induced rat livers increased the efficacy of the exogenous MAS routinely used by FETAX.
- Published
- 2001
- Full Text
- View/download PDF
20. 1,2-Bis(methylsulfonyl)-1-(2-chloroethyl)-2-(methylamino)carbonylhydrazine (101M): a novel sulfonylhydrazine prodrug with broad-spectrum antineoplastic activity.
- Author
-
Finch RA, Shyam K, Penketh PG, and Sartorelli AC
- Subjects
- Animals, Carmustine pharmacology, Drug Screening Assays, Antitumor, Female, Humans, Leukemia L1210 drug therapy, Melanoma, Experimental drug therapy, Mice, Mice, Inbred BALB C, Nitroso Compounds pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Hydrazines pharmacology, Prodrugs pharmacology
- Abstract
Our laboratory has synthesized and evaluated the anticancer activity of a number of sulfonylhydrazine DNA modifying agents. As a class, these compounds possess broad spectrum antitumor activity, demonstrating significant activity against a variety of experimental murine tumors, including the P388 and L1210 leukemias, B16 melanoma, M109 lung carcinoma, and M5076 reticulum cell sarcoma, as well as against the human LX-1 lung carcinoma xenograft. The current report describes the activity of a more recently synthesized member of this class, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-(methylamino)carbonylhydrazine (101M). 101M was active in mice against the i.p. implanted L1210 leukemia over a wide range of doses and produced long-term survivors when administered as a single i.p. bolus of 10, 20, 40, 60, or 80 mg/kg, demonstrating a wider margin of safety than the nitrosourea, 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). Curative therapy was achieved with doses of 101M that did not produce depression of the bone marrow. 101M was also highly effective against the L1210 leukemia when administered by the oral route. The ability of 101M to penetrate the blood-brain barrier and eradicate leukemia cells in the brain was remarkable (>6 log kill). This agent was also curative against L1210 variants resistant to cyclophosphamide, BCNU, or melphalan. Mice implanted with the murine C26 colon carcinoma were also cured by two injections of 10 or 20 mg/kg of 101M. Administration of 101M by two different well-tolerated regimens caused complete regression of established human glioblastoma U251 xenografts in 100% of treated mice, and significant responses were also obtained with 101M against advanced murine M109 lung carcinomas in mice. The broad spectrum of anticancer activity of the sulfonylhydrazine prodrug 101M coupled with the wide range of therapeutic safety exhibited by this agent, makes 101M particularly attractive for further development and clinical evaluation.
- Published
- 2001
21. The pharmacological phenotype of combined multidrug-resistance mdr1a/1b- and mrp1-deficient mice.
- Author
-
Johnson DR, Finch RA, Lin ZP, Zeiss CJ, and Sartorelli AC
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, ATP-Binding Cassette Transporters genetics, Animals, Antineoplastic Agents, Phytogenic pharmacokinetics, Blotting, Western, Crosses, Genetic, Dose-Response Relationship, Drug, Etoposide pharmacokinetics, Female, Male, Mice, Mice, Knockout, Multidrug Resistance-Associated Proteins, Phenotype, Vincristine pharmacokinetics, ATP-Binding Cassette Transporters metabolism, Antineoplastic Agents, Phytogenic toxicity, Drug Resistance, Multiple genetics, Etoposide toxicity, Genes, MDR genetics, Vincristine toxicity
- Abstract
Two major classes of plasma membrane proteins that actively extrude a wide range of structurally diverse hydrophobic amphipathic antineoplastic agents from cells, with different mechanisms of action, lead to multidrug resistance. To study the importance of these ATP-binding cassette transporters to the toxicity of cancer chemotherapy agents, we have used mice genetically deficient in both the mdr1a and mdr1b genes [mdr1a/1b(-/-) mice], the mrp1 gene [mrp1(-/-) mice], and the combined genes mdr1a/1b and mrp1 [mdr1a/1b(-/-), mrp1(-/-) mice] and embryonic fibroblasts derived from wild-type mice and from the three gene knockout animals. The consequences of export pump deficiencies were evaluated primarily using vincristine and etoposide. Mice deficient in the three genes, mdr1a/1b and mrp1, exhibited a 128-fold increase in toxicity to vincristine and a 3-5-fold increase in toxicity to etoposide; increased toxicity to embryonic fibroblast cells from triple knockout mice also occurred with vincristine and etoposide. Vincristine, which normally does not express toxicity to the bone marrow and to the gastrointestinal mucosa when used at therapeutic doses, caused extensive damage to these tissues in mdr1a/1b(-/-), mrp1(-/-) mice. The findings indicate that the P-glycoprotein and mrpl are compensatory transporters for vincristine and etoposide in the bone marrow and the gastrointestinal mucosa and emphasize the potential for increased toxicities by the combined inhibition of these efflux pumps.
- Published
- 2001
22. Evaluation of a reproductive toxicity assay using Xenopus laevis: boric acid, cadmium and ethylene glycol monomethyl ether.
- Author
-
Fort DJ, Stover EL, Bantle JA, Dumont JN, and Finch RA
- Subjects
- Abnormalities, Drug-Induced etiology, Animals, Boric Acids toxicity, Cadmium toxicity, Ethylene Glycols toxicity, Female, Fertility drug effects, Gametogenesis drug effects, Male, Maternal Exposure, Models, Animal, Ovary drug effects, Ovary physiology, Paternal Exposure, Reproduction drug effects, Reproduction physiology, Testis drug effects, Testis physiology, Animal Testing Alternatives, Fertility physiology, Gametogenesis physiology, Xenopus laevis physiology
- Abstract
Cadmium (Cd), boric acid (BA) and ethylene glycol monomethyl ether (EGME) were evaluated for reproductive and developmental toxicity in Xenopus laevis. Eight reproductively mature adult male and eight superovulated female Xenopus laevis were exposed to at least five separate sublethal concentrations of each material via the culture water for a period of 30 days. Four respective pairs were mated and the offspring evaluated for developmental effects; an evaluation of reproductive status was performed on the remaining four specimens. Ovary pathology, oocyte count, oocyte maturity and maturation capacity (germinal vesicle breakdown, GVBD) and necrosis were evaluated in the female, whereas testis pathology, sperm count, dysmorphology and motility were studied in the male. Based on this assessment, each test material exerted reproductive toxicity in Xenopus laevis, but with varying potencies. Adult female exposure to Cd and EGME particularly, and to a lesser extent to BA, resulted in transgenerational toxicity to the developing progeny. Further, this model appears to be a useful tool in the initial assessment and prioritization of potential reproductive toxicants for further testing., (Copyright 2001 John Wiley & Sons, Ltd.)
- Published
- 2001
- Full Text
- View/download PDF
23. The leukotriene C(4) transporter MRP1 regulates CCL19 (MIP-3beta, ELC)-dependent mobilization of dendritic cells to lymph nodes.
- Author
-
Robbiani DF, Finch RA, Jäger D, Muller WA, Sartorelli AC, and Randolph GJ
- Subjects
- ATP-Binding Cassette Transporters physiology, Animals, Blotting, Western, Cell Movement, Cells, Cultured, Chemokine CCL19, Chemokines, CC antagonists & inhibitors, Chemotaxis, Dose-Response Relationship, Drug, Flow Cytometry, Fluorescein-5-isothiocyanate pharmacology, Humans, Immunoblotting, Leukotriene Antagonists pharmacology, Leukotriene D4 metabolism, Ligands, Mice, Mice, Knockout, Multidrug Resistance-Associated Proteins, Propionates pharmacology, Quinolines pharmacology, Receptors, CCR7, Receptors, Chemokine metabolism, Reverse Transcriptase Polymerase Chain Reaction, Skin metabolism, ATP-Binding Cassette Transporters metabolism, Chemokines, CC metabolism, Dendritic Cells metabolism, Leukotriene C4 metabolism, Lymph Nodes metabolism
- Abstract
Adaptive immune responses begin after antigen-bearing dendritic cells (DCs) traffic from peripheral tissues to lymph nodes. Here, we show that DC migration from skin to lymph nodes utilizes the leukotriene C(4) (LTC(4)) transporter multidrug resistance-associated protein 1 (MRP1). DC mobilization from the epidermis and trafficking into lymphatic vessels was greatly reduced in MRP1(-/-) mice, but migration was restored by exogenous cysteinyl leukotrienes LTC(4) or LTD(4). In vitro, these cysteinyl leukotrienes promoted optimal chemotaxis to the chemokine CCL19, but not to other related chemokines. Antagonism of CCL19 in vivo prevented DC migration out of the epidermis. Thus, MRP-1 regulates DC migration to lymph nodes, apparently by transporting LTC(4), which in turn promotes chemotaxis to CCL19 and mobilization of DCs from the epidermis.
- Published
- 2000
- Full Text
- View/download PDF
24. Maintenance of retinoic acid receptor alpha pools by granulocyte colony-stimulating factor and lithium chloride in all-trans retinoic acid-treated WEHI-3B leukemia cells: relevance to the synergistic induction of terminal differentiation.
- Author
-
Finch RA, Li J, Chou TC, and Sartorelli AC
- Subjects
- Antineoplastic Agents therapeutic use, Cell Differentiation drug effects, Drug Synergism, Granulocyte Colony-Stimulating Factor therapeutic use, Humans, Lithium Chloride therapeutic use, Tretinoin therapeutic use, Tumor Cells, Cultured, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Granulocyte Colony-Stimulating Factor pharmacology, Leukemia, Myelomonocytic, Acute drug therapy, Leukemia, Myelomonocytic, Acute metabolism, Leukemia, Myelomonocytic, Acute pathology, Lithium Chloride pharmacology, Receptors, Retinoic Acid metabolism, Tretinoin metabolism, Tretinoin pharmacology
- Abstract
Previous studies have demonstrated that combinations of all-trans retinoic acid (ATRA) with either granulocyte-colony stimulating factor (G-CSF) or lithium chloride (LiCl) produced synergistic terminal differentiation of WEHI-3B myelomonocytic leukemia (D(+)) cells. It was found that steady-state retinoic acid receptor alpha (RARalpha) protein levels were markedly reduced in these cells after exposure to ATRA. Because the presence of receptors for a hormone ligand is required for its action, differentiation therapy with ATRA may be self-limiting. The combination of G-CSF with ATRA significantly attenuated the loss of RARalpha protein, and synergistic terminal differentiation occurred. LiCl was more effective than G-CSF in preserving RARalpha pools and synergized with ATRA more strongly than G-CSF. These findings suggested that the prevention of RARalpha protein loss by G-CSF or LiCl in ATRA-treated cells functioned to extend the differentiation response to the retinoid and was responsible, at least in part, for the observed synergism. D(+) cells transfected with an expression plasmid containing RARalpha cDNA had a 6- to 8-fold increase in steady-state RARalpha mRNA compared with vector-transfected cells and showed a 2- to 3-fold increase in RARalpha protein. ATRA caused a reduction, but not a complete loss, of RARalpha protein in these transfectants, which were considerably more responsive than parental D(+) cells to ATRA as a single agent, supporting the concept that the protection of RARalpha pools results in a heightened differentiation response to ATRA.
- Published
- 2000
25. Triapine (3-aminopyridine-2-carboxaldehyde- thiosemicarbazone): A potent inhibitor of ribonucleotide reductase activity with broad spectrum antitumor activity.
- Author
-
Finch RA, Liu M, Grill SP, Rose WC, Loomis R, Vasquez KM, Cheng Y, and Sartorelli AC
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Blood-Brain Barrier, Cell Division drug effects, DNA biosynthesis, DNA drug effects, Drug Resistance, Neoplasm, Enzyme Inhibitors pharmacokinetics, Enzyme Inhibitors therapeutic use, Humans, Hydroxyurea pharmacology, KB Cells, Leukemia L1210 drug therapy, Mice, Neoplasm Transplantation, Neoplasms, Experimental drug therapy, Pyridines pharmacokinetics, Pyridines therapeutic use, Thiosemicarbazones pharmacokinetics, Thiosemicarbazones therapeutic use, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Pyridines pharmacology, Ribonucleotide Reductases antagonists & inhibitors, Thiosemicarbazones pharmacology
- Abstract
Previous studies from our laboratories have shown that (a) Triapine() is a potent inhibitor of ribonucleotide reductase activity and (b) hydroxyurea-resistant L1210 leukemia cells are fully sensitive to Triapine. In an analogous manner, Triapine was similarly active against the wild-type and a hydroxyurea-resistant subline of the human KB nasopharyngeal carcinoma. Triapine was active in vivo against the L1210 leukemia over a broad range of dosages and was curative for some mice. This agent also caused pronounced inhibition of the growth of the murine M109 lung carcinoma and human A2780 ovarian carcinoma xenografts in mice. Optimum anticancer activity required twice daily dosing due to the duration of inhibition of DNA synthesis which lasted about 10 hr in L1210 cells treated with Triapine in vivo. DNA synthesis in normal mouse tissues (i.e. duodenum and bone marrow) uniformly recovered faster than that in L1210 leukemia cells, demonstrating a pharmacological basis for the therapeutic index of this agent. Triapine was more potent than hydroxyurea in inhibiting DNA synthesis in L1210 cells in vivo, and the effects of Triapine were more pronounced. In addition, the duration of the inhibition of DNA synthesis in leukemia cells from mice treated with Triapine was considerably longer than in those from animals treated with hydroxyurea. Combination of Triapine with various classes of agents that damage DNA (e.g. etoposide, cisplatin, doxorubicin, and 1-acetyl-1,2-bis(methylsulfonyl)-2-(2-chloroethyl)hydrazine) resulted in synergistic inhibition of the L1210 leukemia, producing long-term survivors of tumor-bearing mice treated with several dosage levels of the combinations, whereas no enhancement of survival was found when Triapine was combined with gemcitabine or cytosine arabinoside. The findings demonstrate the superiority of Triapine over hydroxyurea as an anticancer agent and further suggest that prevention by Triapine of repair of DNA lesions created by agents that damage DNA may result in efficacious drug combinations for the treatment of cancer.
- Published
- 2000
- Full Text
- View/download PDF
26. Evaluation of the developmental toxicity of thalidomide using frog embryo teratogenesis assay-xenopus (FETAX): biotransformation and detoxification.
- Author
-
Fort DJ, Stover EL, Bantle JA, and Finch RA
- Subjects
- Amitrole pharmacology, Animals, Benzoflavones pharmacology, Biotransformation, Cyclohexanes pharmacology, Cyclohexenes, Cytochrome P-450 Enzyme System metabolism, Embryo, Nonmammalian metabolism, Embryonic Development, Enzyme Inhibitors pharmacology, Extremities embryology, Extremities pathology, Male, Microsomes, Liver drug effects, Microsomes, Liver enzymology, No-Observed-Adverse-Effect Level, Rats, Rats, Sprague-Dawley, Teratogens metabolism, Thalidomide metabolism, Toxicity Tests methods, Xenopus laevis embryology, Xenopus laevis metabolism, Embryo, Nonmammalian drug effects, Teratogens toxicity, Thalidomide toxicity
- Abstract
The developmental toxicity of thalidomide was evaluated using FETAX (Frog Embryo Teratogenesis Assay - Xenopus). Young X. Laevis embryos were exposed to this compound in each of two concentration-response experiments with and without differently induced exogenous metabolic activation systems (MASs) and/or inhibited MASs. Young male Sprague-Dawley rats were treated with either isoniazid or Aroclor 1254 to induce cytochrome P-450. Several of the rats were subsequently treated with diethyl maleate (DM) to deplete glutathione reserves. Specific aliquots of rat liver microsomes were treated with 3-amino-1,2,4-triazole (ATZ) or alpha-napthoflavone (alpha-N) to selectively inhibit P-450 activity. Bioactivation was indicated by increased developmental toxicity observed in MAS tests. Results obtained indicated that thalidomide was predominantly activated by P-450 isozyne CYP2E1, although weak cross-specificity between CYP1A1/A2 may have existed. Detoxification pathways for thalidomide were investigated by treatment of the MAS with cyclohexene oxide (CHO) and DM to inhibit the epoxide hydrolase and glutathione conjugation pathways, respectively. Results indicated that epoxide hydrolase was primarily responsible for the detoxification of bioactivated thalidomide. Teratogenesis Carcinog. Mutagen. 20:35-47, 2000., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2000
- Full Text
- View/download PDF
27. Phase III interlaboratory study of FETAX. Part 3. FETAX validation using 12 compounds with and without an exogenous metabolic activation system.
- Author
-
Bantle JA, Finch RA, Fort DJ, Stover EL, Hull M, Kumsher-King M, and Gaudet-Hull AM
- Subjects
- Acetates, Acrylamide toxicity, Animals, Arsenites toxicity, Biological Assay, Biotransformation drug effects, Boric Acids toxicity, Bromates toxicity, Chloroacetates, Chlorodiphenyl (54% Chlorine) toxicity, Dichloroacetic Acid toxicity, Embryo, Mammalian metabolism, Ethylene Glycol toxicity, Ethylene Glycols toxicity, Hydrocarbons, Brominated, In Vitro Techniques, Iodoacetates toxicity, Male, Microsomes, Liver drug effects, Microsomes, Liver metabolism, Phthalic Acids toxicity, Polyethylene Glycols toxicity, Rats, Rats, Sprague-Dawley, Reproducibility of Results, Sodium Compounds toxicity, Trichloroacetic Acid toxicity, Xenopus, Embryo, Mammalian drug effects, Embryo, Nonmammalian, Teratogens toxicity
- Abstract
FETAX (Frog Embryo Teratogenesis Assay-Xenopus) is a 96-h whole-embryo developmental toxicity screening assay that can be used in ecotoxicology and in detecting mammalian developmental toxicants when an in vitro metabolic activation system is employed. A standardized American Society for Testing and Materials (ASTM) guide for the conduct of FETAX has been published, along with a companion atlas that helps in embryo staging and in identifying malformations. As part of the ASTM process, an interlaboratory validation study was undertaken to evaluate the repeatability and reliability of FETAX and to evaluate the potential teratogenic hazard of 12 compounds. Three different laboratories participated in the study. All three participating laboratories had extensive experience with the assay. FETAX intralaboratory and interlaboratory variability, as judged by coefficients of variation, were very low. Potential teratogenic hazard was evaluated using two major criteria from FETAX experiments employing metabolic activation systems (MAS). These were the teratogenic index TI (TI = 96-h lc(50)/96-h ec(50) (malformation)) and the minimum concentration that inhibits growth (MCIG). A compound was considered teratogenic by this criterion when the MCIG was significantly different from controls at concentrations below the 30% level of the MAS 96-h lc(50). Based on the results of this and other studies, a decision table was constructed in order to evaluate additional studies. Severity of malformations caused, especially near the MAS 96-h ec(50) (malformation), were also evaluated. Four compounds were non-teratogenic but two compounds were clearly teratogenic. The remaining six compounds were ranked as equivocal teratogens. The results were discussed in light of the difficulty of producing an adequate decision table. FETAX proved to yield repeatable and reliable data as long as care was taken during range-finding and technicians were adequately trained. The MAS was essential in using FETAX to predict developmental hazard in mammals, and still requires further development., (Copyright 1999 John Wiley & Sons, Ltd.)
- Published
- 1999
- Full Text
- View/download PDF
28. New insights into the biology and pharmacology of the multidrug resistance protein (MRP) from gene knockout models.
- Author
-
Rappa G, Finch RA, Sartorelli AC, and Lorico A
- Subjects
- Animals, Antineoplastic Agents, Phytogenic pharmacology, Biological Transport, Carrier Proteins metabolism, Etoposide pharmacology, Glutathione metabolism, Humans, Leukotriene C4 metabolism, Membrane Transport Proteins, Metals, Heavy metabolism, Mice, Mice, Knockout, Potassium Channels metabolism, Drug Resistance, Neoplasm physiology, Genes, MDR
- Abstract
Growing interest in the MRP (multidrug resistance protein) gene stems from its importance in multidrug resistance to chemotherapy, its possible use in gene therapy, and its relationship with the glutathione system. The recent generation of mrp gene knockout models in vitro and in vivo is providing information on the mechanism of action and the physiological function(s) of mrp. The importance of mrp in protection of normal tissues from the toxicity of the anticancer agent etoposide has been established. A total block of mrp has been found to be compatible with life, suggesting that MRP inhibitors can be safely used for treating cancer patients. In some sub-classes of leukocytes, mrp contributes to the transport of leukotriene C4, an endogenous glutathione-S-conjugate. However, the baseline expression of mrp does not appear to contribute to the export of glutathione-S-conjugates of alkylating agents, and thus does not exert a protective role against their toxicity. Besides being capable of exporting certain glutathione-S-conjugates, mrp also catalyzes the co-transport of GSH and drug and, presumably, a presently unknown endogenous metabolite(s).
- Published
- 1999
- Full Text
- View/download PDF
29. Role of vitamin D3 receptor in the synergistic differentiation of WEHI-3B leukemia cells by vitamin D3 and retinoic acid.
- Author
-
Li J, Finch RA, and Sartorelli AC
- Subjects
- Animals, Cell Differentiation drug effects, Dose-Response Relationship, Drug, Drug Combinations, Drug Synergism, Mice, Tumor Cells, Cultured, Leukemia metabolism, Leukemia pathology, Receptors, Calcitriol physiology, Tretinoin pharmacology
- Abstract
WEHI-3B D- cells differentiate in response to 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) but not to all-trans-retinoic acid (RA) or other inducing agents. Combinations of RA with 1,25-(OH)2D3 interact to produce synergistic differentiation of WEHI-3B D- cells. To determine factors involved in the synergistic interaction, expression of the 1,25-(OH)2D3 receptor (VDR) and retinoid receptors, RARalpha and RXRalpha, was measured. No VDR was detected in untreated WEHI-3B D- cells; however, RA and 1,25-(OH)2D3 when used as single agents caused a slight induction of the VDR and in combination produced a marked increase in the VDR. In contrast, no changes in RARalpha and RXRalpha were initiated by these compounds. An RAR-selective agonist combined with 1,25-(OH)2D3 produced synergistic differentiation of WEHI-3B D- cells, whereas an RXR-selective agonist did not. To gain information on the role of the VDR in the synergistic interaction, the VDR gene was transferred into WEHI-3B D+ cells, in which no VDR was detected and no synergism was produced. Expression of the VDR conferred differentiation responsiveness to 1,25-(OH)2D3 in WEHI-3B D+ cells. These findings suggest that (a) induction of VDR expression is a key component in the synergistic differentiation induced by 1,25-(OH)2D3 and RA and (b) RAR and not RXR must be activated for enhanced induction of the VDR and for the synergistic differentiation produced by RA and 1, 25-(OH)2D3., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
30. Animals as sentinels of human health hazards of environmental chemicals.
- Author
-
van der Schalie WH, Gardner HS Jr, Bantle JA, De Rosa CT, Finch RA, Reif JS, Reuter RH, Backer LC, Burger J, Folmar LC, and Stokes WS
- Subjects
- Animals, Biological Assay, Humans, Risk Assessment, Species Specificity, United States, Environmental Exposure adverse effects, Environmental Health, Environmental Monitoring methods, Environmental Pollutants adverse effects, Sentinel Surveillance veterinary
- Abstract
A workshop titled "Using Sentinel Species Data to Address the Potential Human Health Effects of Chemicals in the Environment," sponsored by the U.S. Army Center for Environmental Health Research, the National Center for Environmental Assessment of the EPA, and the Agency for Toxic Substances and Disease Registry, was held to consider the use of sentinel and surrogate animal species data for evaluating the potential human health effects of chemicals in the environment. The workshop took a broad view of the sentinel species concept, and included mammalian and nonmammalian species, companion animals, food animals, fish, amphibians, and other wildlife. Sentinel species data included observations of wild animals in field situations as well as experimental animal data. Workshop participants identified potential applications for sentinel species data derived from monitoring programs or serendipitous observations and explored the potential use of such information in human health hazard and risk assessments and for evaluating causes or mechanisms of effect. Although it is unlikely that sentinel species data will be used as the sole determinative factor in evaluating human health concerns, such data can be useful as for additional weight of evidence in a risk assessment, for providing early warning of situations requiring further study, or for monitoring the course of remedial activities. Attention was given to the factors impeding the application of sentinel species approaches and their acceptance in the scientific and regulatory communities. Workshop participants identified a number of critical research needs and opportunities for interagency collaboration that could help advance the use of sentinel species approaches.
- Published
- 1999
- Full Text
- View/download PDF
31. Choroid plexus epithelial expression of MDR1 P glycoprotein and multidrug resistance-associated protein contribute to the blood-cerebrospinal-fluid drug-permeability barrier.
- Author
-
Rao VV, Dahlheimer JL, Bardgett ME, Snyder AZ, Finch RA, Sartorelli AC, and Piwnica-Worms D
- Subjects
- 3T3 Cells, ATP Binding Cassette Transporter, Subfamily B, Member 1 deficiency, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Animals, Animals, Newborn, Blood-Brain Barrier physiology, Brain anatomy & histology, Brain diagnostic imaging, Cells, Cultured, Choroid Plexus cytology, Epithelial Cells cytology, Epithelial Cells physiology, Humans, KB Cells, Magnetic Resonance Imaging, Mice, Mice, Inbred C57BL, Mice, Knockout, Rats, Rats, Sprague-Dawley, Recombinant Proteins metabolism, Technetium Tc 99m Sestamibi pharmacokinetics, Tomography, Emission-Computed, Single-Photon, Transfection, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Brain physiology, Capillary Permeability, Cerebrospinal Fluid physiology, Choroid Plexus physiology, Drug Resistance, Multiple genetics, Nervous System Physiological Phenomena
- Abstract
The blood-brain barrier and a blood-cerebrospinal-fluid (CSF) barrier function together to isolate the brain from circulating drugs, toxins, and xenobiotics. The blood-CSF drug-permeability barrier is localized to the epithelium of the choroid plexus (CP). However, the molecular mechanisms regulating drug permeability across the CP epithelium are defined poorly. Herein, we describe a drug-permeability barrier in human and rodent CP mediated by epithelial-specific expression of the MDR1 (multidrug resistance) P glycoprotein (Pgp) and the multidrug resistance-associated protein (MRP). Noninvasive single-photon-emission computed tomography with 99mTc-sestamibi, a membrane-permeant radiopharmaceutical whose transport is mediated by both Pgp and MRP, shows a large blood-to-CSF concentration gradient across intact CP epithelium in humans in vivo. In rats, pharmacokinetic analysis with 99mTc-sestamibi determined the concentration gradient to be greater than 100-fold. In membrane fractions of isolated native CP from rat, mouse, and human, the 170-kDa Pgp and 190-kDa MRP are identified readily. Furthermore, the murine proteins are absent in CP isolated from their respective mdr1a/1b(-/-) and mrp(-/-) gene knockout littermates. As determined by immunohistochemical and drug-transport analysis of native CP and polarized epithelial cell cultures derived from neonatal rat CP, Pgp localizes subapically, conferring an apical-to-basal transepithelial permeation barrier to radiolabeled drugs. Conversely, MRP localizes basolaterally, conferring an opposing basal-to-apical drug-permeation barrier. Together, these transporters may coordinate secretion and reabsorption of natural product substrates and therapeutic drugs, including chemotherapeutic agents, antipsychotics, and HIV protease inhibitors, into and out of the central nervous system.
- Published
- 1999
- Full Text
- View/download PDF
32. Triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone; 3-AP): an inhibitor of ribonucleotide reductase with antineoplastic activity.
- Author
-
Finch RA, Liu MC, Cory AH, Cory JG, and Sartorelli AC
- Subjects
- Animals, Cell Division drug effects, DNA, Neoplasm biosynthesis, Drug Resistance, Female, Hydroxyurea pharmacology, In Vitro Techniques, Iron metabolism, Leukemia L1210 drug therapy, Leukemia L1210 metabolism, Mice, Mice, Inbred C3H, Mice, Inbred DBA, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Pyridines pharmacology, Ribonucleotide Reductases antagonists & inhibitors, Thiosemicarbazones pharmacology
- Abstract
The enzyme RR catalyzes the conversion of ribonucleoside diphosphates to their deoxyribonucleotide counterparts. RR is critical for the generation of the cytosine, adenine, and guanine deoxyribonucleotide 5'-triphosphate building blocks of DNA, which are present in cells as exceedingly small intracellular pools. Therefore, interference with the function of RR might well result in an agent with significant antineoplastic activity, particularly against rapidly proliferating tumor cells. HUr is the only inhibitor of RR in clinical usage; this agent, however, is a relatively poor inhibitor of the enzyme and has a short serum half-life. Consequently, HUr is a relatively weak anticancer agent. In an effort to develop a more potent inhibitor of RR with utility as an anticancer agent, we have synthesized 3-AP and demonstrated (a) potent inhibition of L1210 leukemia cells in vitro, (b) curative capacity for mice bearing the L1210 leukemia, (c) marked inhibition of RR, and (d) sensitivity of HUr-resistant cells to 3-AP. These findings collectively demonstrate the clinical potential of 3-AP as an antineoplastic agent.
- Published
- 1999
- Full Text
- View/download PDF
33. Identification of a repressor of the differentiation of WEHI-3B D- leukemia cells.
- Author
-
Li J, Finch RA, Xiao W, and Sartorelli AC
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors, Cell Differentiation genetics, Cell Division, Cell Fusion, Cell Size, Cholecalciferol pharmacology, DNA-Binding Proteins analysis, Diploidy, Gene Expression, Granulocyte Colony-Stimulating Factor pharmacology, Lithium Chloride pharmacology, Mice, RNA, Messenger analysis, RNA, Neoplasm analysis, T-Cell Acute Lymphocytic Leukemia Protein 1, Transfection, Tretinoin pharmacology, Tumor Cells, Cultured, DNA-Binding Proteins genetics, Leukemia, Myeloid genetics, Leukemia, Myeloid pathology, Polyploidy, Proto-Oncogene Proteins, Transcription Factors
- Abstract
The WEHI-3B D+ leukemia is a near-diploid differentiation-competent cell line that undergoes myeloid differentiation in response to retinoic acid. WEHI-3B D- cells, derived from WEHI-3B D+ cells, are near tetraploid and not responsive to the differentiation-inducing properties of the retinoid. To gain information on mechanisms that regulate the maturation of these two cell lines, several multiploid cell lines have been established through fusion of WEHI-3B D+ and WEHI-3B D- cells. Studies with the multiploid cell lines have shown that (a) the cellular growth rate decreases with increased DNA ploidy; (b) near-tetraploid D+/+ cells, obtained by fusing WEHI-3B D+ with WEHI-3B D+ cells, remain differentiation-competent, demonstrating that no direct relationship exists between differentiation competency and DNA ploidy; and (c) near-hexaploid D +/- and D -/+ cells, formed by fusion of WEHI-3B D+ with WEHI-3B D- cells, do not respond to differentiation inducers, suggesting the inhibition of the differentiation machinery of WEHI-3B D+ cells by components from maturation-incompetent WEHI-3B D- cells. The scl transcription factor gene is expressed in WEHI-3B D- cells and is absent in WEHI-3B D+ cells. Overexpression of scl by transfection of scl cDNA in WEHI-3B D+ cells markedly decreased the capacity of retinoic acid to induce differentiation, suggesting that scl functions as a repressor of differentiation in WEHI-3B cell lines.
- Published
- 1998
- Full Text
- View/download PDF
34. Phase III interlaboratory study of FETAX, Part 2: interlaboratory validation of an exogenous metabolic activation system for frog embryo teratogenesis assay--Xenopus (FETAX).
- Author
-
Fort DJ, Stover EL, Bantle JA, Rayburn JR, Hull MA, Finch RA, Burton DT, Turley SD, Dawson DA, Linder G, Buchwalter D, Dumont JN, Kumsher-King M, and Gaudet-Hull AM
- Subjects
- Animals, Biotransformation, Caffeine pharmacokinetics, Cyclophosphamide pharmacokinetics, Lethal Dose 50, Male, Rats, Rats, Sprague-Dawley, Abnormalities, Drug-Induced, Caffeine toxicity, Cyclophosphamide toxicity, Microsomes, Liver metabolism, Xenopus embryology
- Abstract
Interlaboratory validation of an exogenous metabolic activation system (MAS) developed for the alternative, short-term developmental toxicity bioassay, Frog Embryo Teratogenesis Assay-Xenopus (FETAX) was performed with cyclophosphamide and caffeine. Seven study groups within six separate laboratories participated in the study in which three definitive concentration-response experiments were performed with and without the MAS in a side-by-side format for each chemical. Since both chemicals had been previously tested in FETAX, the test concentrations were provided to each laboratory prior to testing. Interlaboratory coefficient of variation (CV) values for unactivated cyclophosphamide (no MAS) were 15%, 15%, 29%, and 25% for the 96-hr LC50, 96-hr EC50 (malformation), Minimum Concentration to Inhibit Growth (MCIG), and Teratogenic Index (TI) values, respectively. Addition of the MAS increased the CV values of each endpoint at least 3.9-fold. Interlaboratory CV values for unactivated caffeine were 31%, 18%, 31%, and 46% for the 96-hr LC50, 96-hr EC50 (malformation), MCIG, and TI values, respectively. Addition of the MAS decreased the CV values of each respective endpoint by at least 1.6-fold. Results indicated that bioactivated toxicants may be prone to greater variability in response amongst laboratories than compounds, which are detoxified. Even though more variability was noted with activated cyclophosphamide, results were within interlaboratory variation expected for other aquatic-based bioassays. Thus, results from these studies warrant the continued use and further refinement of FETAX for alternative developmental toxicity assessment.
- Published
- 1998
- Full Text
- View/download PDF
35. Disruption of the murine MRP (multidrug resistance protein) gene leads to increased sensitivity to etoposide (VP-16) and increased levels of glutathione.
- Author
-
Lorico A, Rappa G, Finch RA, Yang D, Flavell RA, and Sartorelli AC
- Subjects
- Animals, Arsenites toxicity, Bone Marrow pathology, Cell Line, Cell Survival drug effects, Female, Glutamate-Cysteine Ligase metabolism, Male, Mice, Mice, Knockout, Multidrug Resistance-Associated Proteins, Organ Specificity, Sodium Compounds toxicity, Stem Cells, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Antineoplastic Agents toxicity, Drug Resistance, Multiple genetics, Etoposide analogs & derivatives, Etoposide toxicity, Glutathione metabolism, Organophosphorus Compounds toxicity
- Abstract
The mrp (multidrug resistance protein) gene has been associated with the multidrug resistance of cancer cells in vitro and in vivo. To gain information on its physiological role, embryonic stem cells were used to generate mice homozygous for a disruption of the mrp gene, resulting in complete abrogation of mrp expression. No physiological abnormalities were observed, at least up to 4 months of age. Viability, fertility, and a range of histological, hematological, and serum-chemical parameters were similar in mrp(+/+) and mrp(-/-) mice. mrp(-/-) mice displayed an increased sensitivity to etoposide phosphate (2-fold) accompanied by greater bone marrow toxicity, whereas the acute toxicity of sodium arsenite was equivalent in mrp(+/+) and mrp(-/-) mice. Tissue levels of glutathione (GSH) were elevated in breast, lung, heart, kidney, muscle, colon, testes, bone marrow cells, blood mononuclear leukocytes, and blood erythrocytes of mrp(-/-) mice and were unchanged in organs known to express little if any mrp, such as the liver and small intestine. The increase in GSH was not due to an increase in the activity of gamma-glutamylcysteine synthetase, the rate-limiting enzyme for GSH synthesis. The findings demonstrate that mrp is dispensable for development and growth but exerts a role in drug detoxification and GSH metabolism.
- Published
- 1997
36. Structural and functional relationships of toyocamycin on NPM-translocation.
- Author
-
Finch RA, Revankar GR, and Chan PK
- Subjects
- Biological Transport, HeLa Cells, Humans, Structure-Activity Relationship, Antibiotics, Antineoplastic pharmacology, Toyocamycin pharmacology
- Abstract
Toyocamycin is an antitumor antibiotic which has a pyrrolo[2,3-D]pyrimidine aglycone with a -CN substituent on the 5-carbon. Treatment of HeLa cells with toyocamycin induces redistribution of the nuclear phosphoprotein nucleophosmin/B23 (NPM) from nucleoli to nucleoplasm (NPM-translocation) which can be detected by immunofluorescence. NPM-translocation is useful in showing drug effects and in detecting drug-resistant cancer cells. To study which structural features of toyocamycin are important for NPM-translocation, we used toyocamycin analogs in which the 5-position -CN was either deleted (tubercidin) or replaced with a -CONH2 (sangivamycin) or -C(NOH)NH2. HeLa cells were incubated with these analogs for 4 h and assayed for NPM-translocation by immunofluorescence. We found that the analog with the deletion of the -CN group (tubercidin) did not induce translocation while those with replacement of the -CN group with -CONH2 or -C(NOH)NH2 retained the NPM-translocation activity. When these or similar modifications were applied to 7-deazaguanosine, none of the guanosine analogs were effective. These results indicate that modifications at the 5-position of the pyrrolo[2,3-D]pyrimidine ring and a structure similar to adenine rather than guanine are essential for NPM-translocation. Since inhibition of RNA synthesis did not induce NPM-translocation, our results suggest that interference with NPM's binding in nucleoli by these analogs causes NPM-translocation.
- Published
- 1997
37. FETAX interlaboratory validation study: phase III--Part 1 testing.
- Author
-
Bantle JA, Finch RA, Burton DT, Fort DJ, Dawson DA, Linder G, Rayburn JR, Hull M, Kumsher-King M, Gaudet-Hull AM, and Turley SD
- Subjects
- Aminopropionitrile toxicity, Animals, Arsenates toxicity, Ascorbic Acid toxicity, Copper Sulfate toxicity, Dose-Response Relationship, Drug, Embryonic Development, Observer Variation, Sodium Acetate toxicity, Sodium Glutamate toxicity, Embryo, Nonmammalian drug effects, Teratogens toxicity, Xenopus laevis embryology
- Abstract
The Frog Embryo Teratogenesis Assay-Xenopus (FETAX) is a 96-h whole embryo developmental toxicity screening assay that can be used in ecotoxicology and in detecting mammalian developmental toxicants when an in vitro metabolic activation system is employed. A standardized American Society for Testing and Materials (ASTM) guide for the conduct of FETAX has been published, along with a companion atlas that aids in embryo staging and identifying malformations. As part of the ASTM process, a three-phase interlaboratory validation study was undertaken to evaluate the repeatability and reliability of FETAX. Seven different participants collaborated in the study. In Phase I, FETAX proved to be more repeatable and reliable than many bioassays. However, some excessive variation was observed in a few laboratories. An initial lack of assay experience by some technicians caused variation. Phase II showed far less intra- and interlaboratory variability than Phase I. Non-teratogens showed the most consistent results, while more variability was observed for the two teratogens tested. Interlaboratory coefficient of variation values for all endpoints ranged from 7.3 to 54.7. Phase III--Part 1, using coded samples and test concentration ranges selected by each laboratory, showed results similar to Phase I. Analysis of the causes of variation suggested that some technicians judged some embryos to be malformed while others consistently judged similar embryos as normal. Concentration ranges tested by some of the laboratories varied greatly and a new protocol for selecting concentrations for initial testing was written to reduce variation from this source. Testing to date suggests that FETAX is as repeatable and reliable as other standard bioassays.
- Published
- 1996
- Full Text
- View/download PDF
38. ATP depletion affects NPM translocation and exportation of rRNA from nuclei.
- Author
-
Finch RA and Chan PK
- Subjects
- 2,4-Dinitrophenol, Alanine analogs & derivatives, Alanine pharmacology, Antibiotics, Antineoplastic pharmacology, Azides pharmacology, Cell Nucleus drug effects, Dinitrophenols pharmacology, HeLa Cells, Humans, Kinetics, Mycophenolic Acid pharmacology, Nucleophosmin, Ribosomes metabolism, Sodium Azide, Adenosine Triphosphate metabolism, Cell Nucleus metabolism, Nuclear Proteins metabolism, Nucleolus Organizer Region metabolism, RNA, Ribosomal biosynthesis
- Abstract
Nucleophosmin/B23 (NPM) is a nucleolar phosphoprotein which shifts from nucleoli to the nucleoplasm in cells treated with certain cytotoxic agents (NPM-translocation). NPM requires GTP for localization into nucleoli (J. Biol. Chem. 268, 5823-5827, 1993). To understand more about NPM's dynamic localization, the effects of lowering ATP on NPM-translocation and rRNA synthesis were studied. When the ATP level in HeLa cells was reduced by sodium azide, NPM-translocation was blocked. Similar results were obtained when ATP was depleted by other agents, suggesting that ATP depletion was responsible for the blocking of NPM-translocation. It was found that newly synthesized rRNA accumulated in the nuclei during ATP-depletion. Significantly larger than normal nucleoli were also observed. These results indicate that NPM may be involved in the transportation of newly synthesized ribosomes.
- Published
- 1996
- Full Text
- View/download PDF
39. GTP gamma S restores nucleophosmin (NPM) localization to nucleoli of GTP-depleted HeLa cells.
- Author
-
Finch RA, Chang DC, and Chan PK
- Subjects
- Electroporation, Guanosine 5'-O-(3-Thiotriphosphate) metabolism, HeLa Cells, Humans, Hydrolysis, Mycophenolic Acid pharmacology, Nucleophosmin, RNA, Ribosomal biosynthesis, Cell Nucleolus drug effects, Cell Nucleolus metabolism, Guanosine 5'-O-(3-Thiotriphosphate) pharmacology, Guanosine Triphosphate metabolism, Nuclear Proteins metabolism
- Abstract
Previous studies showed that localization of nucleophosmin/B23 (NPM) to nucleoli requires adequate cellular GTP levels (Finch et al., J Biol Chem 268, 5823-5827, 1993). In order to study whether hydrolysis of GTP plays a role in NPM localization, we introduced a nonhydrolyzable GTP analog into HeLa cells. Cells were first depleted of GTP with the IMP dehydrogenase inhibitor, mycophenolic acid (MA), to induce translocation of NPM from the nucleoli to the nucleoplasm. Non-hydrolyzable GTP analogs were then introduced into cells by electroporation. We found that introduction of the non-hydrolyzable analog, GTP gamma S, was effective in restoring NPM localization to nucleoli. Cells incubated in medium containing G-nucleotides without electroporation showed no effect. To reduce the possibility that cells use guanine from degraded nucleotide to supplement GTP pools via salvage pathways, experiments were also performed in the presence of (6-mercaptopurine) 6MP, a competitive inhibitor of the salvage enzyme, HGPRT (hypoxanthine guanine phosphoribosyl transferase), in addition to MA. Under these conditions, introduction of GTP gamma S still effectively restored the localization of NPM into nucleoli. This study demonstrates that electroporation can be used effectively to introduce nucleotides into cultured cells without excessive loss of viability. Our results also indicate that the GTP dependent localization of NPM to the nucleoli may not require GTP hydrolysis.
- Published
- 1995
- Full Text
- View/download PDF
40. Initial interlaboratory validation study of FETAX: phase I testing.
- Author
-
Bantle JA, Burton DT, Dawson DA, Dumont JN, Finch RA, Fort DJ, Linder G, Rayburn JR, Buchwalter D, and Maurice MA
- Subjects
- 6-Aminonicotinamide toxicity, Animals, Hydroxyurea toxicity, In Vitro Techniques, Isoniazid toxicity, Quality Control, Teratogens toxicity, Toxicology methods, Xenopus laevis abnormalities
- Abstract
An interlaboratory validation study was undertaken to evaluate the repeatability and reliability of the Frog Embryo Teratogenesis Assay-Xenopus (FETAX), which is a whole embryo developmental toxicity screening assay. A three-phase experimental program with seven participants was carried out. Phase I was a training and protocol evaluation phase where the identity of the three test materials was known. Hydroxyurea, isoniazid and 6-aminonicotinamide were tested in Phase I. Because the chemicals has been tested previously in FETAX, the same concentrations needed to establish the 96-h median lethal concentration (LC50) and the concentration inducing malformations in 50% of the surviving embryos (EC50) were used by all laboratories. The results of Phase I are presented in this report, and FETAX has proved to be as repeatable and reliable as many other bioassays. Some excess variation was observed in individual laboratories. Some of this variation may have been due to training difficulties. One change in protocol design necessitated by this study was the use of 6-aminonicotinamide as a reference toxicant. While 6-aminonicotinamide provided excellent concentration-response data in most laboratories, the protocol was written too strictly based on historical FETAX data. Phases II and III are currently in progress.
- Published
- 1994
- Full Text
- View/download PDF
41. Nucleolar localization of nucleophosmin/B23 requires GTP.
- Author
-
Finch RA, Revankar GR, and Chan PK
- Subjects
- Biological Transport, HeLa Cells, Humans, IMP Dehydrogenase antagonists & inhibitors, Nucleophosmin, Ribavirin analogs & derivatives, Ribavirin pharmacology, Cell Nucleolus metabolism, Guanosine Triphosphate metabolism, Nuclear Proteins metabolism, Phosphoproteins metabolism
- Abstract
Incubation of HeLa cells with the IMP dehydrogenase inhibitors: ribavirin (100 microM, 4 h), tiazofurin (100 microM, 4 h), selenazofurin (100 microM, 4 h), or mycophenolic acid (10 microM, 4 h) resulted in approximately 70% reduction in cellular GTP pools and shifting of nucleophosmin/B23 from nucleoli to nucleoplasm as detected by immunofluorescence (B23-translocation). Enzyme-linked immunosorbent assay and Western blot assay showed there is no loss or degradation of nucleophosmin/B23 protein during drug treatment. This translocation effect could be prevented by co-incubation with guanosine (100 microM) or reversed by addition of guanosine (100 microM) to the culture medium after B23-translocation had been induced by these inhibitors. Under these conditions of guanosine supplementation, cellular GTP pool concentrations were maintained at the control level. These results indicate that localization of nucleophosmin/B23 into the nucleolus is dependent on the cellular GTP level.
- Published
- 1993
42. Teratogenic assessment of four solvents using the Frog Embryo Teratogenesis Assay--Xenopus (FETAX).
- Author
-
Dresser TH, Rivera ER, Hoffmann FJ, and Finch RA
- Subjects
- Animals, Dimethyl Sulfoxide toxicity, Dioxolanes toxicity, Ethanol toxicity, Female, Formamides toxicity, Male, Teratogens toxicity, Toxicology methods, Xenopus laevis, Abnormalities, Drug-Induced etiology, Embryonic and Fetal Development drug effects, Solvents toxicity
- Abstract
The Frog Embryo Teratogenesis Assay--Xenopus (FETAX) was used to assess the teratogenic potential of four solvents. Embryos of the South African clawed frog, Xenopus laevis, were exposed for 96 h to ethanol, dimethyl sulfoxide (DMSO), formamide or glycerol formal. Exposure groups were maintained using a static renewal system in which the exposure media were changed at 24-h intervals. Survival was monitored at 24-h intervals. Length, as an indicator of growth effects, and developmental malformations were determined at the end of the assay (96 h). Using this information, the 96-h LC50, the 96-h EC50 (Malformation), and the no observable effect levels (NOELs) for mortality, malformation and length were determined for each solvent. The teratogenic index [TI = 96-h LC50/96-h EC50 (Malformation)] also was calculated for each of the solvents. DMSO appeared to be the least toxic or teratogenic solvent examined, with a pooled LC50 of 1.92%, a pooled EC50 (Malformation) of 1.57% and TI values of 1.20 and 1.24 in replicate trials. Formamide appeared to be the most toxic solvent, with a pooled LC50 of 1.04%. Data trends suggested that ethanol was the most teratogenic solvent tested, with a pooled EC50 (Malformation) of 1.04% and TI values of 1.42 and 1.50. The results obtained in the present work for ethanol and DMSO were compared to previously published FETAX results for these two solvents. The present results are in close agreement with these results from other laboratories, thus providing further evidence supporting the interlaboratory reproducibility of FETAX results.
- Published
- 1992
- Full Text
- View/download PDF
43. Studies of actinomycin D induced B23-translocation in P388D1 cells implanted in DBA/2 mice.
- Author
-
Finch RA and Chan PK
- Subjects
- Animals, Dose-Response Relationship, Drug, Female, Fluorescent Antibody Technique, Leukemia P388 pathology, Mice, Mice, Inbred DBA, Neoplasm Transplantation, Nuclear Proteins analysis, Nucleophosmin, Time Factors, Tumor Cells, Cultured drug effects, Dactinomycin pharmacology, Leukemia P388 genetics, Nuclear Proteins genetics, Translocation, Genetic drug effects
- Abstract
Nucleophosmin/B23 is a nucleolar phosphoprotein which redistributes from nucleoli to nucleoplasm (B23-translocation) when cells are exposed to certain anticancer drugs, particularly intercalators. The B23-translocation assay has been demonstrated in cell culture to correlate with drug effects and to detect drug-resistant cells. We now report the effect of actinomycin D on B23-translocation in P388D1 cells implanted in DBA/2 mice. B23-translocation was observed in cells after actinomycin D treatment in a dosage- and time-dependent manner. Translocation could be observed within 30 min after drug treatment. Complete B23-translocation with at least 1-day duration was achieved by a single injection of 0.25 mg/kg. Reduced dosages produced partial B23-translocation with shorter durations. These results indicate that B23-translocation may be useful in monitoring drug effects in animals.
- Published
- 1992
- Full Text
- View/download PDF
44. Chemotherapeutic characterization in mice of 2-amino-9-beta-D-ribofuranosylpurine-6-sulfinamide (sulfinosine), a novel purine nucleoside with unique antitumor properties.
- Author
-
Avery TL, Finch RA, Vasquez KM, Radparvar S, Hanna NB, Revankar GR, and Robins RK
- Subjects
- Animals, Dose-Response Relationship, Drug, Drug Resistance, Female, Guanosine analogs & derivatives, Guanosine therapeutic use, Leukemia L1210 drug therapy, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Thionucleosides therapeutic use, Antineoplastic Agents therapeutic use, Neoplasms, Experimental drug therapy, Purine Nucleosides therapeutic use
- Abstract
In preclinical investigations performed in mice, 2-amino-9-beta-D-ribofuranosyl purine-6-sulfinamide (sulfinosine), a novel derivative of 6-thioguanosine (6TGR), was active against six solid tumors and four strains of experimental leukemia. Sulfinosine penetrated the central nervous system more readily than did 6TGR and, when given repeatedly, was much more effective in the treatment of L1210 leukemia, being curative for some mice. Other findings of major interest to us were the different dosing characteristics of sulfinosine and 6TGR, the divergent efficiencies of the two drugs in generating cellular resistance, and the activity of sulfinosine against experimental leukemias refractory to 6TGR and other experimental or clinically used chemotherapeutic agents. The chemotherapeutic characterization of sulfinosine that evolved from these studies suggests that this agent may have unique properties that deserve clinical consideration. Both the dosing characteristics of the drug and its pronounced activity against thiopurine-resistant experimental leukemia favor the possibility that sulfinosine could be used to advantage in the treatment of human leukemia unresponsive to 6-mercaptopurine or 6-thioguanine.
- Published
- 1990
45. Oxidation of 2-amino-9-beta-D-ribofuranosylpurine-6-sulfenamide to the corresponding 6-sulfonamide facilitates changes in biologic characterization that include activity against thiopurine-refractory experimental leukemia.
- Author
-
Finch RA, Vasquez KM, Hanna NB, Revankar GR, Robins RK, and Avery TL
- Subjects
- Animals, Blood-Brain Barrier, Drug Resistance, Female, Guanosine pharmacokinetics, Guanosine therapeutic use, Mice, Oxidation-Reduction, Structure-Activity Relationship, Thionucleosides pharmacokinetics, Antineoplastic Agents therapeutic use, Guanosine analogs & derivatives, Leukemia L1210 drug therapy, Purine Nucleosides therapeutic use, Sulfonamides therapeutic use, Thionucleosides therapeutic use
- Abstract
Preclinical investigations in vivo revealed unexpected differences in the biological characteristics of 2-amino-9-beta-D-ribofuranosylpurine-6-sulfenamide (sulfenosine, 1) and 2-amino-9-beta-D-ribofuranosylpurine-6-sulfonamide (sulfonosine, 2), two novel but structurally related derivatives of 6-thioguanosine (6TGR). Strikingly, the addition of a fully oxidized sulfur atom at the 6 position of sulfenosine produced a purine derivative (sulfonosine) that was remarkably active against experimental leukemia resistant to treatment with either sulfenosine or 6TGR. This slight structural modification also appeared to influence solubility, scheduling capability, and oral activity as well as penetration of the central nervous system (CNS) and the onset of cellular resistance. These findings underscore the dramatic changes in biologic activity that can be produced by subtle modifications in molecular structure. We trust they may also contribute to the development of improved clinical therapy.
- Published
- 1990
- Full Text
- View/download PDF
46. Synthesis and antitumor evaluation in mice of certain 7-deazapurine (pyrrolo[2,3-d]pyrimidine) and 3-deazapurine (imidazo[4,5-c]pyridine) nucleosides structurally related to sulfenosine, sulfinosine, and sulfonosine.
- Author
-
Ramasamy K, Imamura N, Hanna NB, Finch RA, Avery TL, Robins RK, and Revankar GR
- Subjects
- Animals, Chemical Phenomena, Chemistry, Female, Leukemia L1210 drug therapy, Mice, Purine Nucleosides therapeutic use, Pyrimidine Nucleosides pharmacology, Structure-Activity Relationship, Sulfides therapeutic use, Sulfonamides therapeutic use, Sulfoxides therapeutic use, Antineoplastic Agents chemical synthesis, Purine Nucleosides chemical synthesis, Pyrimidine Nucleosides chemical synthesis, Ribonucleosides therapeutic use
- Abstract
7-Deaza (pyrrolo[2,3-d]pyrimidine) and 3-deaza (imidazo[4,5-c]pyridine) congeners of sulfenosine (5a and 9), sulfinosine (6a and 10), and sulfonosine (7a) have been prepared and evaluated for their antileukemic activity in mice. Amination of 2-amino-7-beta-D-ribofuranosylpyrrolo[2,3-d]pyrimidine-4(3H)-th ion e (4a) and its 2'-deoxy analogue (4c) with a chloramine solution gave the corresponding 4-sulfenamides (5a and 5c, respectively), which on selective oxidation with m-chloroperoxybenzoic acid (MCPBA) gave the respective diastereomeric 2-amino-7-beta-D-ribofuranosyl-pyrrolo[2,3-d]pyrimidine-4-sulfinamide (7-deazasulfinosine, 6a) and its 2'-deoxy derivative (6c). A similar amination of 7-(2-deoxy-beta-D-erythro-pentofuranosyl)pyrrolo[2,3-d]pyrimidine-4(3H)- thione (4b) gave the corresponding 4-sulfenamide derivative (5b). Oxidation of 5b with 1 molar equiv of MCPBA furnished (R,S)-7-(2-deoxy-beta-D-erythro-pentofuranosyl)pyrrolo[2,3-d]pyrimidine- 4- sulfinamide (6b), whereas use of excess of MCPBA afforded the corresponding sulfonamide derivative (7b). Treatment of 3-deaza-6-thioguanosine (8) with a chloramine solution gave 3-deazasulfenosine (6-amino-1-beta-D- ribofuranosylimidazo[4,5-c]pyridine-4-sulfenamide, 9). Controlled oxidation of 9 with MCPBA afforded 3-deazasulfinosine (10). As gauged by increases in the mean postinoculation life spans of L1210 inoculated mice, none of these nucleosides exhibited biologically significant activity (T/C greater than or equal to 125). Even so, antileukemic activity appeared to be influenced, albeit not uniformly, by structural modifications in the base and carbohydrate moieties of sulfenosine and sulfinosine. Thus, while several of the compounds were lacking in cytotoxic activity, eight others (4c, 5a, 5c, 6a, 6b, 7b, 9, and 10) were estimated to have reduced body burdens of viable L1210 cells by 16-77%.
- Published
- 1990
- Full Text
- View/download PDF
47. Changes in diacylglycerol and membrane associated protein kinase C activity reflect the growth status of xenografted human mammary carcinoma treated with 8-Cl-cAMP.
- Author
-
Parandoosh Z, Rubalcava B, Finch RA, Robins RK, and Avery TL
- Subjects
- 8-Bromo Cyclic Adenosine Monophosphate pharmacology, Adult, Animals, Cell Membrane enzymology, Female, Humans, Mammary Neoplasms, Experimental analysis, Mice, Neoplasm Transplantation, Transplantation, Heterologous, 8-Bromo Cyclic Adenosine Monophosphate analogs & derivatives, Antineoplastic Agents pharmacology, Diglycerides analysis, Glycerides analysis, Mammary Neoplasms, Experimental pathology, Protein Kinase C analysis
- Abstract
The intracellular accumulation of cAMP inhibits the growth of transformed cells in vitro and in vivo, and exposure to various cAMP analogs produces similar results. The influence of such analogs on the growth of neoplastic cells in vivo is less well defined, and the relevance of these analogs for the phosphoinositide pathway has not been established. The present report details the inhibition of tumor growth that occurred when human mammary xenografts were treated with 8-Cl-cAMP, the subsequent rebound in tumor growth that occurred when treatment ceased, and the levels of diacylglycerol and membrane-associated protein kinase C activity that characterized tumors in different growth states. Tumor levels of diacylglycerol and particulate PKC activity appeared to be influenced not only by treatment but also by treatment withdrawal. Changes in these entities tended to coincide with tumor growth rate, being relatively suppressed during growth stasis and markedly elevated during periods of rapid growth. The data presented do not establish a causal relationship. Thus, the concomitant changes noted in tumor growth and tumor levels of either diacylglycerol and membrane-associated protein kinase C may only be coincidental. Alternatively, they may indicate that cAMP analogs inhibit tumor growth in vivo by modulating the phosphoinositide pathway.
- Published
- 1990
- Full Text
- View/download PDF
48. 1,2,4-Diazaphosphole nucleosides. Synthesis, structure, and antitumor activity of nucleosides with a lambda 3 phosphorus atom.
- Author
-
Riley TA, Larson SB, Avery TL, Finch RA, and Robins RK
- Subjects
- Animals, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Azides adverse effects, Azides therapeutic use, Chemical Phenomena, Chemistry, IMP Dehydrogenase antagonists & inhibitors, Mice, Models, Molecular, Neoplasms, Experimental drug therapy, Organophosphorus Compounds adverse effects, Organophosphorus Compounds therapeutic use, Structure-Activity Relationship, Survival Analysis, X-Ray Diffraction, Antineoplastic Agents chemical synthesis, Azides chemical synthesis, Nucleosides chemical synthesis, Organophosphorus Compounds chemical synthesis
- Abstract
Glycosylation of 1,2,4 lambda 3-diazaphosphole (4) under Lewis acid catalyzed conditions gave 1-alpha-D-ribofuranosyl-1,2,4 lambda 3-diazaphosphole (5) as the only product. Ethyl 1,2,4 lambda 3-diazaphosphole-3-carboxylate (10) was synthesized by the cyclocondensation of ethyl (chlorophosphinidene)(trimethylsilyl)acetate (8) with (trimethylsilyl)diazomethane and subsequent desilylation with tetra-n-butylammonium fluoride. Reaction of 10 with methanolic ammonia at 80 degrees C gave 1,2,4 lambda 3-diazaphosphole-3-carboxamide. Glycosylation of 10 using trimethylsilyl triflate catalyst followed by ammonlysis gave the ribavirin (1) analogue 1-beta-D-ribofuranosyl-1,2,4 lambda 3-diazaphosphole-3-carboxamide (11). Acetylation of 11 and subsequent treatment with phosphorus pentasulfide gave 2',3',5'-tri-O-acetyl-1-beta-D-ribofuranosyl-1,2,4 lambda 3-diazaphosphole-3- thiocarboxamide (13). Deprotection with methanolic ammonia gave 1-beta-D-ribofuranosyl-1,2,4 lambda 3-diazaphosphole-3-thiocarboxamide (14). Compound 14 gave a 25% increase in life span (ILS) against L1210 in female BDF1 mice. The anomeric configuration and site of glycosylation of 5 and 13 were established by single-crystal X-ray crystallography.
- Published
- 1990
- Full Text
- View/download PDF
49. Synthesis and in vivo antitumor activity of 2-amino-9H-purine-6-sulfenamide, -sulfinamide, and -sulfonamide and related purine ribonucleosides.
- Author
-
Revankar GR, Hanna NB, Imamura N, Lewis AF, Larson SB, Finch RA, Avery TL, and Robins RK
- Subjects
- 2-Aminopurine chemical synthesis, 2-Aminopurine therapeutic use, Animals, Chemical Phenomena, Chemistry, Female, Leukemia L1210 drug therapy, Mice, Molecular Conformation, Molecular Structure, Purine Nucleosides chemical synthesis, Ribonucleosides chemical synthesis, Sulfonamides chemical synthesis, X-Ray Diffraction, 2-Aminopurine analogs & derivatives, Adenine analogs & derivatives, Antineoplastic Agents, Purine Nucleosides therapeutic use, Ribonucleosides therapeutic use, Sulfonamides therapeutic use
- Abstract
A number of 6-sulfenamide, 6-sulfinamide, and 6-sulfonamide derivatives of 2-aminopurine and certain related purine ribonucleosides have been synthesized and evaluated for antileukemic activity in mice. Amination of 6-mercaptopurine ribonucleoside (7a) and 6-thioguanosine (7b) with chloramine solution gave 9-beta-D-ribofuranosylpurine-6-sulfenamide (8a) and 2-amino-9-beta-D-ribofuranosylpurine-6-sulfenamide (sulfenosine, 8b), respectively. Selective oxidation of 8a and 8b with 3-chloroperoxybenzoic acid (MCPBA) gave (R,S)-9-beta-D-ribofuranosylpurine-6-sulfinamide (9a) and (R,S)-2-amino-9-beta-D-ribofuranosylpurine-6-sulfinamide (sulfinosine, 9b), respectively. However, oxidation of 8a and 8b with excess of MCPBA gave 9-beta-D-ribofuranosylpurine-6-sulfonamide (10a) and 2-amino-9-beta-D-ribofuranosylpurine-6-sulfonamide (sulfonosine, 10b), respectively. Similarly, amination of 5'-deoxy-6-thioguanosine (7c) afforded the 6-sulfenamide derivative (8c), which on controlled oxidation gave (R,S)-2-amino-9-(5-deoxy-beta-D-ribofuranosyl)purine-6-sulfinamide (9c) and the corresponding 6-sulfonamide derivative (10c). Treatment of 6-thioguanine (12) with aqueous chloramine solution gave 2-amino-9H-purine-6-sulfenamide (13). Oxidation of 13 with 1 molar equiv of MCPBA afforded (R,S)-2-amino-9H-purine-6-sulfinamide (14), whereas the use of 4 molar equiv of MCPBA furnished 2-amino-9H-purine-6-sulfonamide (15). The resolution of R and S diastereomers of sulfinosine (9b) was accomplished by HPLC techniques. The structures of (R)-9b and 10b were assigned by single-crystal X-ray diffraction studies. (R)-9b exists in the crystal structure in four crystallographically independent conformations. Of the 18 compounds evaluated, 13 exhibited very significant anti-L1210 activity in mice. Sulfenosine (8b) at 22 mg/kg per day X 1 showed a T/C of 170, whereas sulfinosine (9b) at 173 mg/kg per day X 1 showed a T/C of 167 against L1210 leukemia. The 5'-deoxy analogue of sulfinosine (9c) at 104 mg/kg per day also showed a T/C of 172. A single treatment with 8b, 9b, and 9c reduced body burdens of viable L1210 cells by more than 99.8%.
- Published
- 1990
- Full Text
- View/download PDF
50. Treatment of Goodpasture's syndrome with immunosuppression and plasmapheresis.
- Author
-
Finch RA, Rutsky EA, McGowan E, and Wilson CB
- Subjects
- Adolescent, Anti-Glomerular Basement Membrane Disease drug therapy, Anti-Glomerular Basement Membrane Disease immunology, Antibodies, Basement Membrane immunology, Humans, Kidney Glomerulus immunology, Male, Anti-Glomerular Basement Membrane Disease therapy, Cyclophosphamide therapeutic use, Plasmapheresis, Prednisone therapeutic use
- Abstract
A 14-year-old boy with Goodpasture's syndrome induced by anti-glomerular-basement-membrane (gmb) antibody exhibited declining renal function, in association with a progressive increase in the level of serum anti-GBM antibody. Treatment with prednisone, cyclophosphamide, and plasmapheresis was associated with rapid disappearance of the serum anti-GMB antibody and temporary stabilization of renal function.
- Published
- 1979
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.