Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Curr Drug Targets ; 24(8): 648-661, 2023.
Article in English | MEDLINE | ID: mdl-37138489

ABSTRACT

Targeting sodium-dependent glucose transporters (SGLT1 and SGLT2) represents a new class of pharmacotherapy for type 2 diabetes mellitus, a major global health issue with an increasing social and economic burden. Following recent successes in market approvals of SGLT2 inhibitors, the ongoing effort has paved the way for the discovery of novel agents via structure-activity relationship studies, preclinical and clinical testing, including SGLT2 inhibitors, SGLT1/2 dual inhibitors, and selective SGLT1 inhibitors. A growing understanding of the physiology of SGLTs allows drug developers to explore additional cardiovascular and renal protective benefits of these agents in T2DM patients at risk. This review provides an overview of the recent investigational compounds and discusses future perspectives of drug discovery in this area.


Subject(s)
Diabetes Mellitus, Type 2 , Sodium-Glucose Transporter 2 Inhibitors , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/chemically induced , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Hypoglycemic Agents/adverse effects , Sodium-Glucose Transporter 2/therapeutic use , Kidney
2.
Acta Pharmacol Sin ; 41(5): 678-685, 2020 May.
Article in English | MEDLINE | ID: mdl-31937933

ABSTRACT

Fecal microbiota transplantation (FMT) has become an effective strategy to treat metabolic diseases, including type 2 diabetes mellitus (T2DM). We previously reported that the intestinal microbiome had significant difference between individuals with normal glucose tolerance and T2DM in Chinese Kazak ethnic group. In this study, we investigated the effects of transplanted fecal bacteria from Kazaks with normal glucose tolerance (KNGT) in db/db mice. The mice were treated with 0.2 mL of fecal bacteria solution from KNGT daily for 10 weeks. We showed that the fecal bacteria from KNGT successfully colonized in the intestinal tract of db/db mice detected on day 14. In the FMT-treated db/db mice, the levels of fasting blood glucose, postprandial glucose, total cholesterol, triglyceride, and low-density lipoprotein-cholesterol were significantly downregulated, whereas high-density lipoprotein-cholesterol levels were upregulated. In the FMT-treated db/db mice, Desulfovibrio and Clostridium coccoides levels in gut were significantly decreased, but the fecal levels of Akkermansia muciniphila and colon histone deacetylase-3 (HDAC3) protein expression were increased. At 8 weeks, both intestinal target bacteria and HDAC3 were correlated with glycolipid levels; Akkermansia muciniphila level was positively correlated with HDAC3 protein expression (r = +0.620, P = 0.037). Our results suggest that fecal bacteria from KNGT could potentially be used to treat diabetic patients.


Subject(s)
Clostridiales/metabolism , Desulfovibrio/metabolism , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Akkermansia/metabolism , Animals , Diabetes Mellitus, Type 2/therapy , Disease Models, Animal , Dyslipidemias/therapy , Humans , Male , Mice
3.
J Inorg Biochem ; 177: 249-258, 2017 12.
Article in English | MEDLINE | ID: mdl-28551160

ABSTRACT

Oxaliplatin-based chemotherapy is the mainstay for the treatment of advanced colorectal cancer. Copper transporter proteins have been implicated in the transport of platinum-based anticancer drugs, but their expression in human colorectal cancer cell lines and roles in controlling their sensitivity to oxaliplatin are not well studied or understood. The endogenous and modified expression of copper uptake transporter 1 (hCTR1) was studied in a panel of human colorectal cancer cell lines (DLD-1, SW620, HCT-15 and COLO205) with ~20-fold variation in oxaliplatin sensitivity. hCTR1 protein was expressed more abundantly than ATP7A and ATP7B proteins, but with broadly similar levels and patterns of expression across four colorectal cancer cell lines. In a colorectal cancer cell-line background (DLD-1), stable transfection of the hCtr1 gene enhanced hCTR1 protein expression and increased the sensitivity of the cells to the cytotoxicity of copper and oxaliplatin. Treatment with copper chelators (ammonium tetrathiomolybdate, bathocuproinedisulfonic acid and D-penicillamine) increased expression of hCTR1 protein in DLD-1 and SW620 cells, and potentiated the cytotoxicity of oxaliplatin in DLD-1 but not SW620 cells. Treatment with copper chloride altered neither the expression of copper transporters nor cytotoxicity of oxaliplatin in colorectal cancer lines. In conclusion, human colorectal cancer cell lines consistently express hCTR1 protein despite their variable sensitivity to oxaliplatin. Genetic or pharmacological modification of hCTR1 protein expression may potentiate oxaliplatin sensitivity in some but not all colorectal cancer cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Cation Transport Proteins/genetics , Organoplatinum Compounds/pharmacology , Cation Transport Proteins/metabolism , Cell Line, Tumor , Chelating Agents/pharmacology , Copper/metabolism , Copper Transporter 1 , Copper-Transporting ATPases/metabolism , Drug Synergism , Humans , Molybdenum/pharmacology , Organoplatinum Compounds/metabolism , Oxaliplatin , Penicillamine/pharmacology , Phenanthrolines/pharmacology , Up-Regulation/drug effects
4.
Oncotarget ; 7(41): 67277-67287, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27572309

ABSTRACT

NVP-BKM120 (BKM120) is a new pan-class I phosphatidylinositol-3 kinase (PI3K) inhibitor and has been tested in clinical trials as an anticancer agent. In this study, we determined whether BKM120 induces autophagy and the impact of autophagy induction on BKM120's growth-inhibitory activity. BKM120 potently induced elevation of autophagosome-bound type II LC3 (LC3-II) protein, predominantly in cell lines insensitive to BKM120, thereby inducing autophagy. The presence of lysosomal protease inhibitor chloroquine further enhanced the levels of LC3-II. BKM120 combined with chloroquine, enhanced growth-inhibitory effects including induction of apoptosis, suggesting that autophagy is a protective mechanism counteracting BKM120's growth-inhibitory activity. Interestingly, BKM120 increased p-ERK1/2 levels. When blocking the activation of this signaling with MEK inhibitors or with knockdown of ERK1/2, the ability of BKM120 to increase LC3-II was attenuated and the growth-inhibitory effects including induction of apoptosis were accordingly enhanced, suggesting that the MEK/ERK activation contributes to BKM120-induced authophagy. In mouse xenograft model, we also found that the combination of BKM120 and PD0325901 synergistically suppressed cell growth in human lung cancer cells. Thus, the current study not only reveals mechanisms accounting for BKM120-induced autophagy, but also suggests an alternative method to enhance BKM120's therapeutic efficacy against non-small cell lung cancer(NSCLC) by blocking autophagy with either a lysosomal protease inhibitor or MEK inhibitor.


Subject(s)
Aminopyridines/pharmacology , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chloroquine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Mice , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Tumour Biol ; 37(8): 11177-86, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26935060

ABSTRACT

MicroRNAs (miRNAs) play a critical role in cancer development and progression. Deregulated expression of miR-204 has been reported in several cancers, but the mechanism through which miR-204 modulates human non-small cell lung cancer (NSCLC) is largely unknown. In this study, we investigate the expression and functional role of miR-204 in human NSCLC tissues and cell lines. RNA isolation, qRT-PCR, MTT, colony formation assay, cell cycle assay, cell apoptosis assay, cell migration assay, and Western blot were performed. Statistical analysis was performed using SPSS 18.0 software and statistical significance was accepted at p value <0.05. miR-204 level was significantly reduced in NSCLC tissues as compared to that of non-neoplastic tissues. Transient over-expression of miR-204 by transfecting with miR-204 mimics suppressed NSCLC cell proliferation, migration, and induced apoptosis and G1 arrest, whereas inhibition of miR-204 showed the converse effects. Additionally, activating transcription factor 2 (ATF2), an important transcription factor, was demonstrated as a potential target gene of miR-204. Subsequent investigations found a negative correlation between miR-204 level and ATF2 expression in NSCLC tissue samples. Moreover, we observed that miR-204 expression inversely affected endogenous ATF2 expression at both mRNA and protein levels in vitro. Taken together, miR-204 may act as a tumor suppressor by directly targeting ATF2 in NSCLC.


Subject(s)
Activating Transcription Factor 2/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Activating Transcription Factor 2/genetics , Adult , Aged , Apoptosis/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Transfection
6.
Sci Rep ; 5: 17348, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26615818

ABSTRACT

Here, we investigated the impact of mulberry fruit (MBF) extracts on lipopolysaccharide (LPS)-induced inflammatory responses in RAW 264.7 macrophages, and the therapeutic efficacy of MBF diet in mice with dextran sulfate sodium (DSS)-induced acute colitis and MUC2(-/-) mice with colorectal cancer. In vitro, LPS-induced nitric oxide (NO) production was significantly inhibited by MBF extracts via suppressing the expression of proinflammatory molecules, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-1 beta (IL-ß) and IL-6. Particularly, a dose-dependent inhibition on LPS-induced inflammatory responses was observed following treatment with MBF dichloromethane extract (MBF-DE), in which linoleic acid and ethyl linolenate were identified as two active compounds. Moreover, we elucidated that MBF-DE attenuated LPS-induced inflammatory responses by blocking activation of both NF-κB/p65 and pERK/MAPK pathways. In vivo, DSS-induced acute colitis was significantly ameliorated in MBF-fed mice as gauged by weight loss, colon morphology and histological damage. In addition, MBF-fed MUC2(-/-) mice displayed significant decrease in intestinal tumor and inflammation incidence compared to control diet-fed group. Overall, our results demonstrated that MBF suppressed the development of intestinal inflammation and tumorgenesis both in vitro and in vivo, and supports the potential of MBF as a therapeutic functional food for testing in human clinical trials.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fruit/chemistry , Macrophages/drug effects , Macrophages/metabolism , Mitogen-Activated Protein Kinases/metabolism , Morus/chemistry , NF-kappa B/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colitis/drug therapy , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/adverse effects , Dietary Supplements , Disease Models, Animal , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Linoleic Acid/chemistry , Linolenic Acids/chemistry , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Mucin-2/deficiency , Nitric Oxide/biosynthesis , Phosphorylation , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Protein Transport
7.
J Neurochem ; 135(6): 1099-112, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364854

ABSTRACT

Platinum-based anticancer drugs cause peripheral neurotoxicity by damaging sensory neurons within the dorsal root ganglia (DRG), but the mechanisms are incompletely understood. The roles of platinum DNA binding, transcription inhibition and altered cell size were investigated in primary cultures of rat DRG cells. Click chemistry quantitative fluorescence imaging of RNA-incorporated 5-ethynyluridine showed high, but wide ranging, global levels of transcription in individual neurons that correlated with their cell body size. Treatment with platinum drugs reduced neuronal transcription and cell body size to an extent that corresponded to the amount of preceding platinum DNA binding, but without any loss of neuronal cells. The effects of platinum drugs on neuronal transcription and cell body size were inhibited by blocking platinum DNA binding with sodium thiosulfate, and mimicked by treatment with a model transcriptional inhibitor, actinomycin D. In vivo oxaliplatin treatment depleted the total RNA content of DRG tissue concurrently with altering DRG neuronal size. These findings point to a mechanism of chemotherapy-induced peripheral neurotoxicity, whereby platinum DNA damage induces global transcriptional arrest leading in turn to neuronal atrophy. DRG neurons may be particularly vulnerable to this mechanism of toxicity because of their requirements for high basal levels of global transcriptional activity. Findings point to a new stepwise mechanism of chemotherapy-induced peripheral neurotoxicity, whereby platinum DNA damage induces global transcriptional arrest leading in turn to neuronal atrophy. Dorsal root ganglion neurons may be particularly vulnerable to this neurotoxicity because of their high global transcriptional outputs, demonstrated in this study by click chemistry quantitative fluorescence imaging.


Subject(s)
DNA Damage/drug effects , DNA/drug effects , Drug Therapy , Neurons/drug effects , Neurotoxicity Syndromes/drug therapy , Organoplatinum Compounds/pharmacology , Platinum/pharmacology , Animals , Cation Transport Proteins/drug effects , Cation Transport Proteins/metabolism , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Oxaliplatin , Rats
8.
Dis Markers ; 2015: 824304, 2015.
Article in English | MEDLINE | ID: mdl-25999661

ABSTRACT

The role of genetics in progression of cancer is an established fact, and susceptibility risk and difference in outcome to chemotherapy may be caused by the variation in low-penetrance alleles of risk genes. We selected seven genes (CRP, GPC5, ACTA2, AGPHD1, SEC14L5, RBMS3, and GKN1) that previously reported link to lung cancer (LC) and genotyped single nucleotide polymorphisms (SNPs) of these genes in a case-control study. A protective allele "C" was found in rs2808630 of the C-reactive protein (CRP). Model association analysis found genotypes "T/C" and "C/C" in the dominant model and genotype "T/C" in the overdominant model of rs2808630 associated with reduced LC risk. Gender-specific analysis in each model showed that genotypes "T/T" and "C/C" in rs2352028 of the Glypican 5 (GPC5) were associated with increased LC risk in males. Logistic regression analysis showed "C/T" genotype carriers of rs4254535 in the Gastrokine 1 (GKN1) had less likelihood to have chemotherapy response. Our results suggest a potential association between CRP and GPC5 variants with LC risk; variation in GKN1 is associated with chemotherapy response in the Chinese Han population.


Subject(s)
Asian People/genetics , Glypicans/genetics , Lung Neoplasms/genetics , Peptide Hormones/genetics , Polymorphism, Single Nucleotide , Vesicular Transport Proteins/genetics , Adult , Asian People/ethnology , Case-Control Studies , China/ethnology , Cisplatin/therapeutic use , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Logistic Models , Lung Neoplasms/drug therapy , Lung Neoplasms/ethnology , Male , Middle Aged
9.
Curr Drug Targets ; 16(12): 1356-71, 2015.
Article in English | MEDLINE | ID: mdl-25901528

ABSTRACT

The superfamily of human ATP-binding cassette (ABC) transporters comprises seven subfamilies (ABCA to G) with 48 members. In addition to their profound physiological and pharmacological functions, ABC transporters play important roles in instigating multidrug resistance (MDR) in cancer by mediating the efflux of many anticancer drugs, particularly, ABCB1, ABCG2 and ABCC subfamily members. Previous development of ABCB1 transporter inhibitors has provided insights into seeking novel strategies in developing new classes of compound that inhibit ABCB1 and other MDRrelated ABC transporters. We herein review and evaluate current evidence in this area, with an emphasis on experimental and investigational agents that are under preclinical and clinical tests, including tyrosine kinase inhibitors, natural products, microRNAs and novel chemical entities. New strategies targeting ABC transporters in cancer stem cells and future perspectives in this field are also discussed.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Animals , Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Humans , MicroRNAs/therapeutic use , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors
10.
Eur J Cancer Prev ; 23(6): 497-501, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25254308

ABSTRACT

Lung cancer has the highest mortality rate among cancers; however, its nosogenesis is still unclear. Genome-wide association studies have shown that the telomerase reverse transcriptase (TERT) gene, located in the chromosome 5p15.33 region, is one of the genes associated with the risk of lung cancer. In this case-control study, we genotyped 11 tag single-nucleotide polymorphisms of the TERT gene to evaluate their association with lung cancer risk in the Han Chinese population. Two tag single-nucleotide polymorphisms were found to be associated with lung cancer risk on using the χ2-test: rs4246742 [odds ratio (OR)=0.77, 95% confidence interval (CI) 0.60-0.98; P=0.03] and rs2853672 (OR=1.26, 95% CI 1.01-1.57; P=0.045). By using SNPStats software we also found rs2242652 (OR=1.47, 95% CI 1.02-2.13; P=0.04) in the dominant model and rs2736098 (OR=1.38, 95% CI 1.06-1.80; P=0.017), rs2853672 (OR=1.41, 95% CI 1.11-1.80; P=0.0048), and rs4246742 (OR=0.75, 95% CI 0.58-0.97; P=0.029) in the log-additive model. 'T/C-T/T' of rs10069690 conferred an increased risk for male sex in the dominant model (OR=1.80, 95% CI, 1.05-3.08; P=0.03) and 'TC' increased risk for male sex in the overdominant model (OR=1.85, 95% CI, 1.08-3.17; P=0.031). Our findings, combined with previous studies, suggest that polymorphisms in the TERT gene contribute to the risk for lung cancer in the Chinese Han population.


Subject(s)
Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Telomerase/genetics , Adult , Aged , Asian People/genetics , Case-Control Studies , China/epidemiology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lung Neoplasms/epidemiology , Male , Middle Aged , Risk Factors
11.
Clin Exp Pharmacol Physiol ; 40(6): 371-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23556474

ABSTRACT

The purpose of the present study was to determine whether copper histidine could inhibit copper transporter 1 (Ctr1)-mediated transport of oxaliplatin in vitro and thereby limit the accumulation of platinum and neurotoxicity of oxaliplatin in dorsal root ganglion (DRG) tissue in vivo. In HEK293 cells overexpressing rat Ctr1, copper histidine was shown to be transported by Ctr1 and to inhibit their Ctr1-mediated uptake of oxaliplatin. Pilot in vivo dose-finding studies showed that copper histidine at doses up to 2 mg/kg, p.o., daily for 5 days/week could be added to maximum tolerated doses of oxaliplatin (1.85 mg/kg, i.p., twice weekly) for 8 week combination treatment studies in female Wistar rats. After treatment, rats showed significant changes in sensory neuron size profiles in DRG tissue induced by oxaliplatin that were not altered by its coadministration with copper histidine. The expression of copper transporters (Ctr1 and copper-transporting P-type ATPase 1 (Atp7a)) in DRG tissue appeared unchanged following treatment with oxaliplatin given alone or with copper histidine. Platinum and copper tissue levels were higher in DRG than in most other tissues, but were unaltered by the addition of copper histidine to oxaliplatin treatment. In conclusion, copper histidine inhibited cellular uptake of oxaliplatin mediated by Ctr1 in vitro without altering the accumulation of platinum or neurotoxicity of oxaliplatin in DRG tissue in vivo at doses tolerated in combination with oxaliplatin treatment.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Ganglia, Spinal/drug effects , Heavy Metal Poisoning, Nervous System/prevention & control , Histidine/analogs & derivatives , Organometallic Compounds/pharmacology , Organoplatinum Compounds/toxicity , Platinum/metabolism , Adenosine Triphosphatases/metabolism , Animals , Body Weight/drug effects , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/metabolism , Copper Transporter 1 , Copper-Transporting ATPases , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Interactions , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HEK293 Cells , Heavy Metal Poisoning, Nervous System/pathology , Histidine/pharmacology , Humans , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin , Rats , Transfection
12.
Biochem Pharmacol ; 85(2): 207-15, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23123662

ABSTRACT

Dorsal root ganglion (DRG) neurons are affected by platinum-induced neurotoxicity and neurodegenerative processes associated with disturbed copper homeostasis and transport. This study aimed to understand the role of copper transporter 1 (Ctr1) in the uptake and toxicity of copper and platinum drugs in cultured rat DRG neurons, and the functional activities of rat Ctr1 (rCtr1) as a membrane transporter of copper and platinum drugs. Heterologous expression of rCtr1 in HEK293 cells (HEK/rCtr1 cells) increased the uptake and cytotoxicity of copper, oxaliplatin, cisplatin and carboplatin, in comparison to isogenic vector-transfected control cells. Cultured rat DRG neurons endogenously expressed rCtr1 protein on their neuronal cell body plasma membranes and cytoplasm, and displayed substantial capacity for taking up copper, but were resistant to copper toxicity. The uptake of copper by both cultured rat DRG neurons and HEK/rCtr1 cells was saturable and inhibited by cold temperature, silver and zinc, consistent with it being mediated by rCtr1. Cultured rat DRG neurons accumulated platinum during their exposure to oxaliplatin and were sensitive to oxaliplatin cytotoxicity. The accumulation of platinum by both cultured rat DRG neurons and HEK/rCtr1 cells, during oxaliplatin exposure, was saturable and temperature dependent, but was inhibited by copper only in HEK/rCtr1 cells. In conclusion, rCtr1 can transport copper and platinum drugs, and sensitizes cells to their cytotoxicities. DRG neurons display substantial capacity for accumulating copper via a transport process mediated by rCtr1, but appear able to resist copper toxicity and use alternative mechanisms to take up oxaliplatin.


Subject(s)
Antineoplastic Agents/metabolism , Cation Transport Proteins/metabolism , Copper/adverse effects , Ganglia, Spinal/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organoplatinum Compounds/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Carboplatin/adverse effects , Carboplatin/metabolism , Carboplatin/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cells, Cultured , Cisplatin/adverse effects , Cisplatin/metabolism , Cisplatin/pharmacology , Copper/metabolism , Copper/pharmacology , Copper Sulfate/adverse effects , Copper Sulfate/metabolism , Copper Sulfate/pharmacology , Copper Transporter 1 , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Membrane Transport Modulators/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/pharmacology , Oxaliplatin , Rats , Rats, Wistar , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
13.
Curr Cancer Drug Targets ; 12(8): 962-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22794121

ABSTRACT

Membrane transporters govern the movement of drugs and their metabolites across biological membranes, thereby determining their pharmacokinetics, efficacy and adverse drug reactions. Platinum-based anticancer drugs are a mainstay of chemotherapy for many human malignancies. However, their clinical utility is limited by tumor resistance and normal tissue toxicities, which are determined at least in part by the level of tissue accumulation of platinum. Recently, several members of the ATP-binding cassette (ABC), solute carrier (SLC) and ATPase membrane protein superfamilies have been found to contribute to the net accumulation of platinum drugs in malignant and normal tissues. Herein, a review has been carried out to critically evaluate current preclinical and clinical evidence implicating membrane transporters as determinants of the pharmacology of cisplatin, oxaliplatin, carboplatin and related investigational compounds. The evidence includes studies of recombinant cell systems with genetically modified expression of individual membrane transporters, platinum-resistant or -sensitive human cancer cells and in vivo xenografted tumors, animal models of platinum-induced nephro-, oto- or neurotoxicity, and clinical studies of associations between the membrane transporter tumor expression and patient outcomes from platinum-based chemotherapy. Understanding the role of membrane transporters as determinants of the pharmacology of platinum drugs will be a basis for targeting these drug transporters in individualized and optimized platinum-based cancer therapy, and new drug development.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Membrane Transport Proteins/metabolism , Platinum Compounds/pharmacology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cisplatin/pharmacology , Cisplatin/toxicity , Copper-Transporting ATPases , Humans , Membrane Transport Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Platinum/pharmacology , Platinum/therapeutic use , Platinum/toxicity , Platinum Compounds/pharmacokinetics , SLC31 Proteins , Treatment Outcome
14.
J Pharmacol Exp Ther ; 338(2): 537-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21606177

ABSTRACT

The organic cation/carnitine transporters OCTN1 and OCTN2 are related to other organic cation transporters (OCT1, OCT2, and OCT3) known for transporting oxaliplatin, an anticancer drug with dose-limiting neurotoxicity. In this study, we sought to determine whether OCTN1 and OCTN2 also transported oxaliplatin and to characterize their functional expression and contributions to its neuronal accumulation and neurotoxicity in dorsal root ganglion (DRG) neurons relative to those of OCTs. [(14)C]Oxaliplatin uptake, platinum accumulation, and cytotoxicity were determined in OCTN-overexpressing human embryonic kidney (HEK) 293 cells and primary cultures of rat DRG neurons. Levels of mRNA and functional activities of rat (r)Octns and rOcts in rat DRG tissue and primary cultures were characterized using reverse transcription-polymerase chain reaction and uptake of model OCT/OCTN substrates, including [(3)H]1-methyl-4-phenylpyridinium (MPP(+)) (OCT1-3), [(14)C]tetraethylammonium bromide (TEA(+)) (OCT1-3 and OCTN1/2), [(3)H]ergothioneine (OCTN1), and [(3)H]l-carnitine (OCTN2). HEK293 cells overexpressing rOctn1, rOctn2, human OCTN1, and human OCTN2 showed increased uptake and cytotoxicity of oxaliplatin compared with mock-transfected HEK293 controls; in addition, both uptake and cytotoxicity were inhibited by ergothioneine and L-carnitine. The uptake of ergothioneine mediated by OCTN1 and of L-carnitine mediated by OCTN2 was decreased during oxaliplatin exposure. rOctn1 and rOctn2 mRNA was readily detected in rat DRG tissue, and they were functionally active in cultured rat DRG neurons, more so than rOct1, rOct2, or rOct3. DRG neuronal accumulation of [(14)C]oxaliplatin and platinum during oxaliplatin exposure depended on time, concentration, temperature, and sodium and was inhibited by ergothioneine and to a lesser extent by L-carnitine but not by MPP(+). Loss of DRG neuronal viability during oxaliplatin exposure was inhibited by ergothioneine but not by L-carnitine or MPP(+). OCTN1 and OCTN2 both transport oxaliplatin and are functionally expressed by DRG neurons. OCTN1-mediated transport of oxaliplatin appears to contribute to its neuronal accumulation and treatment-limiting neurotoxicity more so than OCTN2 or OCTs.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression Regulation , Neurons/metabolism , Organic Cation Transport Proteins/physiology , Organoplatinum Compounds/metabolism , Animals , Biological Transport, Active/genetics , Cells, Cultured , Female , HEK293 Cells , Humans , Organic Cation Transport Proteins/biosynthesis , Organic Cation Transport Proteins/genetics , Oxaliplatin , Rats , Rats, Wistar , Solute Carrier Family 22 Member 5 , Symporters
15.
Mol Pain ; 6: 53, 2010 Sep 13.
Article in English | MEDLINE | ID: mdl-20836889

ABSTRACT

BACKGROUND: ATP7A, ATP7B and CTR1 are metal transporting proteins that control the cellular disposition of copper and platinum drugs, but their expression in dorsal root ganglion (DRG) tissue and their role in platinum-induced neurotoxicity are unknown. To investigate the DRG expression of ATP7A, ATP7B and CTR1, lumbar DRG and reference tissues were collected for real time quantitative PCR, RT-PCR, immunohistochemistry and Western blot analysis from healthy control adult rats or from animals treated with intraperitoneal oxaliplatin (1.85 mg/kg) or drug vehicle twice weekly for 8 weeks. RESULTS: In DRG tissue from healthy control animals, ATP7A mRNA was clearly detectable at levels similar to those found in the brain and spinal cord, and intense ATP7A immunoreactivity was localised to the cytoplasm of cell bodies of smaller DRG neurons without staining of satellite cells, nerve fibres or co-localisation with phosphorylated heavy neurofilament subunit (pNF-H). High levels of CTR1 mRNA were detected in all tissues from healthy control animals, and strong CTR1 immunoreactivity was associated with plasma membranes and vesicular cytoplasmic structures of the cell bodies of larger-sized DRG neurons without co-localization with ATP7A. DRG neurons with strong expression of ATP7A or CTR1 had distinct cell body size profiles with minimal overlap between them. Oxaliplatin treatment did not alter the size profile of strongly ATP7A-immunoreactive neurons but significantly reduced the size profile of strongly CTR1-immunoreactive neurons. ATP7B mRNA was barely detectable, and no specific immunoreactivity for ATP7B was found, in DRG tissue from healthy control animals. CONCLUSIONS: In conclusion, adult rat DRG tissue exhibits a specific pattern of expression of copper transporters with distinct subsets of peripheral sensory neurons intensely expressing either ATP7A or CTR1, but not both or ATP7B. The neuron subtype-specific and largely non-overlapping distribution of ATP7A and CTR1 within rat DRG tissue may be required to support the potentially differing cuproenzyme requirements of distinct subsets of sensory neurons, and could influence the transport and neurotoxicity of oxaliplatin.


Subject(s)
Adenosine Triphosphatases/genetics , Aging/metabolism , Cation Transport Proteins/genetics , Ganglia, Spinal/metabolism , Gene Expression Regulation , Adenosine Triphosphatases/metabolism , Aging/drug effects , Animals , Cation Transport Proteins/metabolism , Cell Size/drug effects , Copper Transporter 1 , Copper-Transporting ATPases , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Gene Expression Regulation/drug effects , Lumbar Vertebrae/cytology , Lumbar Vertebrae/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Pain ; 5: 66, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19922644

ABSTRACT

BACKGROUND: Oxaliplatin and related chemotherapeutic drugs cause painful chronic peripheral neuropathies in cancer patients. We investigated changes in neuronal size profiles and neurofilament immunoreactivity in L5 dorsal root ganglion (DRG) tissue of adult female Wistar rats after multiple-dose treatment with oxaliplatin, cisplatin, carboplatin or paclitaxel. RESULTS: After treatment with oxaliplatin, phosphorylated neurofilament heavy subunit (pNF-H) immunoreactivity was reduced in neuronal cell bodies, but unchanged in nerve fibres, of the L5 DRG. Morphometric analysis confirmed significant changes in the number (-75%; P < 0.0002) and size (-45%; P < 0.0001) of pNF-H-immunoreactive neurons after oxaliplatin treatment. pNF-H-immunoreactive neurons had overlapping size profiles and co-localisation with neurons displaying cell body immunoreactivity for parvalbumin, non-phospho-specific neurofilament medium subunit (NF-M) and non-phospho-specific neurofilament heavy subunit (NF-H), in control DRG. However, there were no significant changes in the numbers of neurons with immunoreactivity for parvalbumin (4.6%, P = 0.82), NF-M (-1%, P = 0.96) or NF-H (0%; P = 0.93) after oxaliplatin treatment, although the sizes of parvalbumin (-29%, P = 0.047), NF-M (-11%, P = 0.038) and NF-H (-28%; P = 0.0033) immunoreactive neurons were reduced. In an independent comparison of different chemotherapeutic agents, the number of pNF-H-immunoreactive neurons was significantly altered by oxaliplatin (-77.2%; P < 0.0001) and cisplatin (-35.2%; P = 0.03) but not by carboplatin or paclitaxel, and their mean cell body area was significantly changed by oxaliplatin (-31.1%; P = 0.008) but not by cisplatin, carboplatin or paclitaxel. CONCLUSION: This study has demonstrated a specific pattern of loss of pNF-H immunoreactivity in rat DRG tissue that corresponds with the relative neurotoxicity of oxaliplatin, cisplatin and carboplatin. Loss of pNF-H may be mechanistically linked to oxaliplatin-induced neuronal atrophy, and serves as a readily measureable endpoint of its neurotoxicity in the rat model.


Subject(s)
Antineoplastic Agents/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Neurofilament Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/adverse effects , Carboplatin/adverse effects , Carboplatin/pharmacology , Cisplatin/adverse effects , Cisplatin/pharmacology , Female , Immunohistochemistry , Organoplatinum Compounds/adverse effects , Oxaliplatin , Paclitaxel/adverse effects , Paclitaxel/pharmacology , Rats , Rats, Wistar
17.
Cancer Chemother Pharmacol ; 64(4): 847-56, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19466412

ABSTRACT

PURPOSE: We report the neuronal expression of copper transporter 1 (CTR1) in rat dorsal root ganglia (DRG) and its association with the neurotoxicity of platinum-based drugs. METHODS: CTR1 expression was studied by immunohistochemistry and RT-PCR. The toxicity of platinum drugs to CTR1-positive and CTR1-negative neurons was compared in DRG from animals treated with maximum tolerated doses of oxaliplatin (1.85 mg/kg), cisplatin (1 mg/kg) or carboplatin (8 mg/kg) twice weekly for 8 weeks. RESULTS: Abundant CTR1 mRNA was detected in DRG tissue. CTR1 immunoreactivity was associated with plasma membranes and cytoplasmic vesicular structures of a subpopulation (13.6 +/- 3.1%) of mainly large-sized (mean cell body area, 1,787 +/- 127 microm(2)) DRG neurons. After treatment with platinum drugs, the cell bodies of these CTR1-positive neurons became atrophied, with oxaliplatin causing the greatest percentage reduction in the mean cell body area relative to controls (42%; P < 0.05), followed by cisplatin (18%; P < 0.05) and carboplatin causing the least reduction (3.2%; P = NS). CTR1-negative neurons, with no immunoreactivity or only diffuse cytoplasmic staining, showed less treatment-induced cell body atrophy than CTR1-positive neurons. CONCLUSIONS: CTR1 is preferentially expressed by a subset of DRG neurons that are particularly vulnerable to the toxicity of platinum drugs. These findings, together with its neuronal membrane localization, are suggestive of CTR1-related mechanisms of platinum drug neuronal uptake and neurotoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cation Transport Proteins/metabolism , Ganglia, Spinal/drug effects , Neurons/drug effects , Organoplatinum Compounds/toxicity , Animals , Cation Transport Proteins/genetics , Copper Transporter 1 , Ganglia, Spinal/metabolism , Immunohistochemistry , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
Clin Exp Pharmacol Physiol ; 35(12): 1440-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18671714

ABSTRACT

1. Satraplatin is an investigational orally administered platinum-based antitumour drug. The present study compared the plasma protein binding, stability and degradation of satraplatin with that of its active metabolite JM118 and cisplatin. 2. The platinum complexes were incubated in human plasma for up to 2 h at 37 degrees C and quantified in plasma fractions by inductively coupled plasma-mass spectrometry on- or off-line to high-performance liquid chromatography. 3. All three platinum drugs became irreversibly bound to plasma proteins and showed negligible reversible protein binding. They were also unstable in plasma and generated one or more platinum-containing degradation products during their incubation. However, the three platinum complexes differed in the kinetics of their instability and protein binding, as well as in the number of degradation products formed during their incubation. 4. In conclusion, the plasma protein binding, instability and degradation of satraplatin and its active metabolite JM118 are qualitatively similar to that of cisplatin and other clinically approved platinum-based drugs. Quantitative differences in their irreversible protein binding and degradation were related to their respective physiochemical properties and bioactivation mechanisms.


Subject(s)
Antineoplastic Agents/blood , Cisplatin/blood , Drugs, Investigational/metabolism , Organoplatinum Compounds/blood , Antineoplastic Agents/metabolism , Cisplatin/metabolism , Drug Stability , Humans , Organoplatinum Compounds/metabolism , Protein Binding/physiology
19.
J Inorg Biochem ; 102(2): 303-10, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18029019

ABSTRACT

In this study we characterised the in vitro antitumour and hepatotoxicity profiles of a series of Au(I) and Ag(I) bidentate phenyl and pyridyl complexes in a panel of cisplatin-resistant human ovarian cancer cell-lines, and in isolated rat hepatocytes. The gold and silver compounds overcame cisplatin-resistance in the CH1-cisR, 41M-cisR and SKOV-3 cell-lines, and showed cytotoxic potencies strongly correlated with their lipophilicity. Complexes with phenyl or 2-pyridyl ligands had high antitumour and hepatotoxic potency and low selectivity between different cell-lines. Their cytotoxicity profiles were similar to classic mitochondrial poisons and an example of this type of compound was shown to accumulate preferentially in the mitochondria of cancer cells in a manner that depended upon the mitochondrial membrane potential. In contrast, complexes with 3- or 4-pyridyl ligands had low antitumour and hepatotoxic potency and cytotoxicity profiles similar to 2-deoxy-D-glucose. In addition, they showed high selectivity between different cell-lines that was not attributable to variation in uptake in different cell-types. The in vitro hepatotoxic potency of the series of gold and silver compounds varied by over 61-fold and was closely related to their lipophilicity and hepatocyte uptake. In conclusion, Au(I) and Ag(I) bidendate pyridyl phosphine complexes demonstrate activity against cisplatin-resistant human cancer cells and in vitro cytotoxicity that strongly depends upon their lipophilicity.


Subject(s)
Antineoplastic Agents/pharmacology , Hepatocytes/drug effects , Organogold Compounds/pharmacology , Organometallic Compounds/pharmacology , Phosphines/pharmacology , Silver/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cisplatin/pharmacology , Hepatocytes/metabolism , Humans , Male , Organogold Compounds/chemistry , Organogold Compounds/toxicity , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , Phosphines/chemistry , Phosphines/toxicity , Rats , Rats, Wistar , Silver/chemistry , Silver/toxicity
20.
Cancer Chemother Pharmacol ; 59(5): 661-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16944150

ABSTRACT

PURPOSE: To evaluate the antitumour activity of 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a vascular disrupting agent currently under phase II clinical trials in combination with cancer chemotherapy, in rats bearing chemically induced primary mammary tumours. METHODS: Tumours were induced in female Wistar rats by injection of N-nitroso-N-methylurea at 100 mg/kg subcutaneously. A clinically relevant single dose of DMXAA (1,800 mg/m(2)) was given to animals when tumours were measurable. Tumour volume, extent of necrosis and cytokine profiles were measured. RESULTS: Compared with the control group, DMXAA treatment significantly delayed tumour doubling time and extended the time from treatment to euthanasia. Four of five DMXAA-treated animals showed necrosis involving 3.7-41.2% of the area of the tumour section at 24 h compared with none of four control animals (P < 0.028, Chi-square test). Intratumoural levels of TNFalpha, IL-6, VEGF and IL-1alpha were increased 4 h after DMXAA treatment. CONCLUSIONS: This study shows for the first time that DMXAA has significant in vivo antitumour activity against non-transplanted autochthonous tumours and in a host species other than the mouse.


Subject(s)
Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Xanthones/therapeutic use , Animals , Apoptosis/drug effects , Carcinogens , Cytokines/biosynthesis , Female , Immunoassay , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...