Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Musculoskelet Neuronal Interact ; 24(2): 178-184, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38826000

ABSTRACT

OBJECTIVE: To investigate the effect of pericapsular nerve group (PENG) block combined with spinal anesthesia in the treatment of elderly patients with intertrochanteric fractures through "rapid diagnosis and treatment channel" PFNA internal fixation. METHODS: 52 elderly patients were randomly divided into the observation group (26 patients, PENG block combined with spinal anesthesia) and the control group (26 patients, spinal anesthesia alone). The general health, mean arterial pressure (MAP), and heart rate (HR) of both groups were compared at various stages: immediately before the administration of pain analgesia, during the positioning of spinal epidural anesthesia, at the beginning and end of the surgery, and 2 hours after surgery. Additionally, VAS scores at rest and during passive straight leg elevation by 15° were evaluated at 12 hours, 24 hours, 48 hours, 72 hours, and 7 days after surgery. RESULTS: The MAP and HR in the observation group under spinal anesthesia in the lateral position were lower than those in the control group (P < 0.05). Additionally, the VAS scores of the observation group during positioning and at 12 hours and 24 hours after surgery were lower than those in the control group under spinal epidural anesthesia (both P < 0.05). CONCLUSION: The application of ultrasound-guided PENG block combined with lumbar anesthesia can reduce pain when in lateral position, stabilize perioperative vital signs, and result in high satisfaction.


Subject(s)
Anesthesia, Spinal , Hip Fractures , Nerve Block , Humans , Anesthesia, Spinal/methods , Aged , Male , Female , Nerve Block/methods , Hip Fractures/surgery , Aged, 80 and over , Pain, Postoperative/prevention & control , Pain, Postoperative/drug therapy
2.
J Cell Mol Med ; 24(22): 13336-13345, 2020 11.
Article in English | MEDLINE | ID: mdl-33030301

ABSTRACT

Aster tataricus L.f. is a traditional Eastern Asian herbal medicine used for the relief of uroschesis-related illnesses and has been demonstrated clinically to exert satisfied effects. However, the mechanism of its therapeutic action remains unclear. The present study aimed to evaluate the protective mechanism of Aster tataricus extract (ATE) on CYP or LPS + ATP-induced interstitial cystitis (IC), we successfully constructed the induced IC Sprague-Dawley (SD) rat model and IC human urothelium cell (SV-HUC-1) model. The main compounds of ATE were determined by LC-MS. After intervention, the changes on the bladder wall morphology and inflammation were observed in each group. SV-HUC1 cell viability was measured by MTT and double stained with Hoechst 33342 and propidium iodide (PI). The expression levels of NLRP3, Pro-caspase-1, Caspsae-1 p20, GSDMD, GSDMD-N and Cleave-IL-1ß in vivo and in vitro in different groups were detected by Western blotting. ATE significantly alleviated oedema and haemorrhage and reduced the inflammation index and histopathological score in SD rat bladder. The results of cell revealed that ATE could improve cell viability and decrease pyroptosis ratio. The expression of NLRP3 and other pyroptosis-related protein was remarkably decreased by ATE both in vivo and in vitro. ATE may be used as an inhibitor of NLRP3 in treating IC. The discovery of NLRP3/Caspase-1/GSDMD-N as a new protective pathway provides a new direction for protecting cell against IC.


Subject(s)
Magnoliopsida/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plant Extracts/pharmacology , Pyroptosis/drug effects , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Cell Survival/drug effects , Chromatography, Liquid , Cystitis/drug therapy , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Humans , Inflammasomes , Inflammation/pathology , Mass Spectrometry , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Urinary Bladder/cytology , Urothelium/cytology
3.
Nucleic Acids Res ; 43(19): 9350-61, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26433227

ABSTRACT

Antisense oligonucleotides (ASOs) are known to trigger mRNA degradation in the nucleus via an RNase H-dependent mechanism. We have now identified a putative cytoplasmic mechanism through which ASO gapmers silence their targets when transfected or delivered gymnotically (i.e. in the absence of any transfection reagent). We have shown that the ASO gapmers can interact with the Ago-2 PAZ domain and can localize into GW-182 mRNA-degradation bodies (GW-bodies). The degradation products of the targeted mRNA, however, are not generated by Ago-2-directed cleavage. The apparent identification of a cytoplasmic pathway complements the previously known nuclear activity of ASOs and concurrently suggests that nuclear localization is not an absolute requirement for gene silencing.


Subject(s)
Cytoplasm/metabolism , Gene Silencing , Oligonucleotides, Antisense , Argonaute Proteins/metabolism , Cell Line , Cytoplasm/chemistry , Gene Transfer Techniques , Oligonucleotides, Antisense/analysis , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Transfection
4.
J Alzheimers Dis ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38905043

ABSTRACT

Background: Alzheimer's disease (AD) is the most common sort of neurodegenerative dementia, characterized by its challenging, diverse, and progressive nature. Despite significant progress in neuroscience, the current treatment strategies remain suboptimal. Objective: Identifying a more accurate molecular target for the involvement of microglia in the pathogenic process of AD and exploring potential mechanisms via which it could influence disease. Methods: We utilized single-cell RNA sequencing (scRNA-seq) analysis in conjunction with APP/PS1 mouse models to find out the molecular mechanism of AD. With the goal of investigating the cellular heterogeneity of AD, we downloaded the scRNA-seq data from the Gene Expression Omnibus (GEO) database and identified differentially expressed genes (DEGs). Additionally, we evaluated learning and memory capacity using the behavioral experiment. We also examined the expression of proteins associated with memory using western blotting. Immunofluorescence was employed to investigate alterations in amyloid plaques and microglia. Results: Our findings revealed an upregulation of ITGAX expression in APP/PS1 transgenic mice, which coincided with a downregulation of synaptic plasticity-related proteins, an increase in amyloid-ß (Aß) plaques, and an elevation in the number of M1 microglia. Interestingly, deletion of ITGAX resulted in increased Aß plaque deposition, a rise in the M1 microglial phenotype, and decreased production of synaptic plasticity-related proteins, all of which contributed to a decline in learning and memory. Conclusions: This research suggested that ITGAX may have a beneficial impact on the APP/PS1 mice model, as its decreased expression could exacerbate the impairment of synaptic plasticity and worsen cognitive dysfunction.

5.
J Mol Neurosci ; 74(2): 42, 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38613644

ABSTRACT

Alzheimer's disease (AD) is a severe neurological illness that causes memory loss and is a global problem. The calcium hypothesis recently steadily evolved in AD. The prospective targets for calcium homeostasis therapy, however, are limited, and gene expression-level research connected to calcium homeostasis in AD remains hazy. In this study, we analyzed the microarray dataset (GSE132903) taken from the Gene Expression Omnibus (GEO) database to investigate calcium homeostasis-related genes for AD. Using immunoblot analysis, we examined the association of ITPKB with inflammation in AD. Additionally, the immunofluorescence technique was employed to assess the impact of pharmacological inhibition of ITPKB on the amyloid-ß (Aß) plaque deposition in APP/PS1 mice. This article's further exploration of calcium homeostasis-related genes has propelled the validation of the calcium homeostasis theory in AD.


Subject(s)
Alzheimer Disease , Plaque, Amyloid , Animals , Mice , Plaque, Amyloid/genetics , Transcriptome , Calcium , Alzheimer Disease/genetics , Models, Animal , Homeostasis
6.
J Pain Res ; 17: 1651-1661, 2024.
Article in English | MEDLINE | ID: mdl-38736679

ABSTRACT

Purpose: Patients undergoing arthroscopic hip surgery (AHS) require good analgesia and early rehabilitation after surgery, and there is no consensus on the optimal nerve block. We aimed to compare the efficacy of the pericapsular nerve group (PENG) block with lateral femoral cutaneous nerve (LFCN) block compared to fascia iliaca compartment block (FICB) in patients with AHS. Patients and Methods: A total of 80 patients receiving AHS under general anesthesia were randomized to receive either FICB (group F) or PENG block in combination with LFCN block (group P). The primary outcomes were the rate of quadriceps weakness after block on the afflicted side, as well as muscle strength grading and pain score after block, and the quality of recovery on the second postoperative day. Results: Compared with group F, group P had a lower incidence of quadriceps weakness 48 h after block (76.9% vs 28.2%, P < 0.001), and had less impact on muscle strength grade and lower static pain score at 6, 12, 18, 24, 36, and 48 h after block (P < 0.001), and a lower dynamic pain score at 6 and 12 h after block in group P (p < 0.05). The quality of recovery on the second postoperative day improved (p < 0.05). Conclusion: In comparison to FICB, PENG block in combination with LFCN block can affect less quadriceps muscle strength and reduce the use of postoperative analgesics, which is beneficial for the postoperative recovery of AHS patients.

7.
Hum Gene Ther ; 33(1-2): 37-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34806402

ABSTRACT

Huntington's disease is a fatal neurodegenerative disorder caused by an inherited mutation in the huntingtin (HTT) gene comprising an expanded cytosine-adenine-guanine (CAG) trinucleotide repeat sequence that results in a pathogenic huntingtin protein. Adeno-associated viral (AAV) gene therapy containing a primary artificial microRNA (pri-amiRNA) specifically targeting HTT messenger RNA (mRNA) has the potential to provide long-lasting therapeutic benefit, through durable reduction of mutant HTT expression after a single administration. The efficiency and precision of processing of the pri-amiRNA precursor to the mature guide (G) strand by transduced cells are critical for specific and potent HTT mRNA lowering. The selection of the optimized pri-amiRNA comprised a series of in vitro studies followed by in vivo studies in small and then large mammals. Our studies demonstrate the predictivity of certain cell culture systems and rodent models for nonhuman primates with respect to some, but not all key features of pri-amiRNA processing. In addition, our results show that the processing of pri-amiRNAs to the mature guide strand can differ greatly across different scaffolds and sequences while providing the same levels of target lowering. Importantly, our data demonstrate that there is a combinatorial effect of guide and passenger (P) strand sequences, together with the scaffold, on pri-amiRNA processing, with different guide and passenger strand sequences within the same scaffold dramatically altering pri-amiRNA processing. Taken together, our results highlight the importance of optimizing not only target lowering but also the efficiency and precision of pri-amiRNA processing in vitro, in rodents and in large mammals to identify the most potent and selective AAV gene therapy that harnesses the endogenous microRNA (miRNA) biogenesis pathway for target lowering without perturbing the endogenous cellular miRNA profile. The optimized pri-amiRNA was selected with this focus on efficiency and precision of pri-amiRNA processing in addition to its pharmacological activity on HTT mRNA lowering and general tolerability in vivo.


Subject(s)
Huntington Disease , MicroRNAs , Animals , Genetic Therapy , Genetic Vectors/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/therapy , Mice , MicroRNAs/genetics , Primates/genetics
8.
J Clin Invest ; 118(6): 2246-59, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18464931

ABSTRACT

Long QT syndrome (LQTS) is a heritable disease associated with ECG QT interval prolongation, ventricular tachycardia, and sudden cardiac death in young patients. Among genotyped individuals, mutations in genes encoding repolarizing K+ channels (LQT1:KCNQ1; LQT2:KCNH2) are present in approximately 90% of affected individuals. Expression of pore mutants of the human genes KCNQ1 (KvLQT1-Y315S) and KCNH2 (HERG-G628S) in the rabbit heart produced transgenic rabbits with a long QT phenotype. Prolongations of QT intervals and action potential durations were due to the elimination of IKs and IKr currents in cardiomyocytes. LQT2 rabbits showed a high incidence of spontaneous sudden cardiac death (>50% at 1 year) due to polymorphic ventricular tachycardia. Optical mapping revealed increased spatial dispersion of repolarization underlying the arrhythmias. Both transgenes caused downregulation of the remaining complementary IKr and IKs without affecting the steady state levels of the native polypeptides. Thus, the elimination of 1 repolarizing current was associated with downregulation of the reciprocal repolarizing current rather than with the compensatory upregulation observed previously in LQTS mouse models. This suggests that mutant KvLQT1 and HERG interacted with the reciprocal wild-type alpha subunits of rabbit ERG and KvLQT1, respectively. These results have implications for understanding the nature and heterogeneity of cardiac arrhythmias and sudden cardiac death.


Subject(s)
KCNQ1 Potassium Channel/genetics , Long QT Syndrome/genetics , Long QT Syndrome/pathology , Action Potentials , Animals , Animals, Genetically Modified , Death, Sudden , Disease Models, Animal , ERG1 Potassium Channel , Echocardiography , Electrophysiology/methods , Ether-A-Go-Go Potassium Channels , Genotype , Heart Ventricles/pathology , Muscle Cells/pathology , Phenotype , Potassium Channels, Voltage-Gated/genetics , Rabbits
9.
Mol Ther Methods Clin Dev ; 20: 366-378, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33553485

ABSTRACT

Therapeutic payload delivery to the central nervous system (CNS) remains a major challenge in gene therapy. Recent studies using function-driven evolution of adeno-associated virus (AAV) vectors have successfully identified engineered capsids with improved blood-brain barrier (BBB) penetration and CNS tropism in mouse. However, these strategies require transgenic animals and thus are limited to rodents. To address this issue, we developed a directed evolution approach based on recovery of capsid library RNA transcribed from CNS-restricted promoters. This RNA-driven screen platform, termed TRACER (Tropism Redirection of AAV by Cell-type-specific Expression of RNA), was tested in the mouse with AAV9 peptide display libraries and showed rapid emergence of dominant sequences. Ten individual variants were characterized and showed up to 400-fold higher brain transduction over AAV9 following systemic administration. Our results demonstrate that the TRACER platform allows rapid selection of AAV capsids with robust BBB penetration and CNS tropism in non-transgenic animals.

10.
Am J Physiol Heart Circ Physiol ; 299(5): H1525-34, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833965

ABSTRACT

We previously reported a transgenic rabbit model of long QT syndrome based on overexpression of pore mutants of repolarizing K(+) channels KvLQT1 (LQT1) and HERG (LQT2).The transgenes in these rabbits eliminated the slow and fast components of the delayed rectifier K(+) current (I(Ks) and I(Kr), respectively), as expected. Interestingly, the expressed pore mutants of HERG and KvLQT1 downregulated the remaining reciprocal repolarizing currents, I(Ks) and I(Kr), without affecting the steady-state levels of the native polypeptides. Here, we sought to further explore the functional interactions between HERG and KvLQT1 in heterologous expression systems. Stable Chinese hamster ovary (CHO) cell lines expressing KvLQT1-minK or HERG were transiently transfected with expression vectors coding for mutant or wild-type HERG or KvLQT1. Transiently expressed pore mutant or wild-type KvLQT1 downregulated I(Kr) in HERG stable CHO cell lines by 70% and 44%, respectively. Immunostaining revealed a severalfold lower surface expression of HERG, which could account for the reduction in I(Kr) upon KvLQT1 expression. Deletion of the KvLQT1 NH(2)-terminus did not abolish the downregulation, suggesting that the interactions between the two channels are mediated through their COOH-termini. Similarly, transiently expressed HERG reduced I(Ks) in KvLQT1-minK stable cells. Coimmunoprecipitations indicated a direct interaction between HERG and KvLQT1, and surface plasmon resonance analysis demonstrated a specific, physical association between the COOH-termini of KvLQT1 and HERG. Here, we present an in vitro model system consistent with the in vivo reciprocal downregulation of repolarizing currents seen in transgenic rabbit models, illustrating the importance of the transfection method when studying heterologous ion channel expression and trafficking. Moreover, our data suggest that interactions between KvLQT1 and HERG are mediated through COOH-termini.


Subject(s)
Action Potentials/physiology , Down-Regulation/physiology , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/physiology , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/physiology , Mutation/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Down-Regulation/genetics , ERG1 Potassium Channel , Electrophysiology , Female , Gene Deletion , Humans , Kidney/cytology , Kidney/physiology , Ovary/cytology , Ovary/physiology , Patch-Clamp Techniques , Transfection
11.
J Neurosci ; 25(28): 6676-86, 2005 Jul 13.
Article in English | MEDLINE | ID: mdl-16014729

ABSTRACT

The structural determinants of nicotinic acetylcholine receptor (AChR) trafficking have yet to be fully elucidated. Hydrophobic residues occur within short motifs important for endoplasmic reticulum (ER) export or endocytotic trafficking. Hence, we tested whether highly conserved hydrophobic residues, primarily leucines, in the cytoplasmic domain of the alpha4beta2 AChR subunits were required for cell surface expression of alpha4beta2 AChRs. Mutation of F350, L351, L357, and L358 to alanine in the alpha4 AChR subunit attenuates cell surface expression of mutant alpha4beta2 AChRs. Mutation of F342, L343, L349, and L350 to alanine at homologous positions in the beta2 AChR subunit abolishes cell surface expression of mutant alpha4beta2 AChRs. The hydrophobic nature of the leucine residue is a primary determinant of its function because mutation of L343 to another hydrophobic amino acid, phenylalanine, in the beta2 AChR subunit only poorly inhibits trafficking of mutant alpha4beta2 AChR to the cell surface. All mutant alpha4beta2 AChRs exhibit high-affinity binding for [3H]epibatidine. In both tsA201 cells and differentiated SH-SY5Y neural cells, wild-type alpha4beta2 AChRs colocalize with the Golgi marker giantin, whereas mutant alpha4beta2 AChRs fail to do so. The striking difference between mutant alpha4 versus mutant beta2 AChR subunits on cell surface expression of mutant alpha4beta2 AChRs points to a cooperative or regulatory role for the alpha4 AChR subunit and an obligatory role for the beta2 AChR subunit in ER export. Collectively, our results identify, for the first time, residues within AChR subunits that are essential structural determinants of alpha4beta2 AChR ER export.


Subject(s)
Endoplasmic Reticulum/metabolism , Receptors, Nicotinic/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cell Line , Cell Membrane/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kidney , Leucine/chemistry , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuroblastoma/pathology , Neurons/cytology , Neurons/metabolism , Protein Multimerization , Protein Structure, Tertiary , Protein Transport , Pyridines/metabolism , Rats , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , Tumor Cells, Cultured
12.
J Biochem ; 140(3): 313-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16861249

ABSTRACT

Multidrug resistance protein 1 (MRP1) is a human ATP-binding cassette (ABC) transporter in the plasma membrane. It confers multidrug resistance to tumor cells by actively effluxing intracellular drugs. To examine the functional significance of intracellular loops (ICLs) in MRP1, we determined the effect of mutation of the amino acid sequence EXXXG, which is conserved in ICL5 and ICL7 of human MRP1, 2 and 3, sulfonylurea receptor (SUR) 1 and 2, and mouse MRP1 and 2. E and G in the ICLs of human MRP1 were mutated to L and P, respectively, and the N-terminal (including ICL5) and C-terminal (including ICL7) wild type or mutant halves of MRP1 were co-expressed in insect cells. The mutation of either ICL5 or ICL7 considerably decreased ATP-dependent LTC4 uptake into vesicles of insect cells expressing mutated MRP1. GSH-dependent photolabeling of MRP1 with an 125I-labeled photoaffinity analog of azido agosterol A (azido AG-A) was abolished by the mutations in ICL5 and ICL7. Mutations in ICL5 of MRP1 almost completely inhibited the labeling of NBD2, but not NBD1, by 8-azido-alpha-[32P]ATP. In contrast, mutations in ICL7 of MRP1 abolished the labeling of both NBDs. Mutation of either ICL5 or ICL7 of MRP1 almost completely inhibited vanadate trapping with 8-azido-alpha-[32P]ATP by both NBD1 and NBD2 domains. These findings indicate that the intramolecular signaling between NBD and ICLs in MRP1 is vital for MRP1 function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Multidrug Resistance-Associated Proteins/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , DNA Primers , Humans , Leukotriene C4/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Photoaffinity Labels , Protein Structure, Tertiary , Species Specificity , Transport Vesicles/metabolism , Tritium
13.
Cancer Res ; 63(14): 4048-54, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12874005

ABSTRACT

ABCG2 is a plasma membrane efflux pump that is able to confer resistance to several anticancer agents, including mitoxantrone, camptothecins, anthracyclines, and flavopiridol. The antimetabolite methotrexate (MTX) was inferred recently to be an additional substrate of the pump based on the analysis of ABCG2-overexpressing cell lines. However, the transport characteristics of the pump with regard to this agent have not been determined. In addition, physiological substrates of ABCG2 have not been identified. Here we examine the in vitro transport properties of the pump using membrane vesicles prepared from HEK293 cells transfected with ABCG2 expression vector. In so doing it is shown that MTX is a high capacity low affinity substrate of the pump, with K(m) and V(max) values of 1.34 +/- 0.18 mM and 687 +/- 87 pmol/mg/min, respectively. Unlike previously characterized multidrug resistance protein family members, ABCG2 is also able to transport MTX diglutamate and MTX triglutamate. However, addition of even one more glutamyl residue is sufficient to completely abrogate ABCG2-mediated transport. By contrast with the wild-type protein (ABCG2-R482), two ABCG2 variants that have been identified in drug selected cell lines, R482T and R482G, were unable to transport MTX to any extent. Similarly, folic acid was subject to efflux by the wild-type protein but not by the two mutants. However, transport of the reduced folate leucovorin was not detected for either the wild-type or the mutant proteins. Finally, it is shown that ABCG2 is capable of transporting E(2)17betaG with K(m) and V(max) values of 44.2 +/- 4.3 micro M and 103 +/- 17 pmol/mg/min, respectively. These results indicate that ABCG2 is a component of the energy-dependent efflux system for certain folates and antifolates, but that its transport characteristics with respect to polyglutamates and reduced folates are not identical to those of multidrug resistance protein family members. In addition, it is demonstrated that R482 mutations observed in drug-resistant cell lines have profound effects on the in vitro transport properties of the pump.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , Methotrexate/analogs & derivatives , Methotrexate/pharmacokinetics , Mutation , Neoplasm Proteins , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Biological Transport/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Indoles/pharmacology , Transfection
14.
Biochem Pharmacol ; 70(7): 1056-65, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16098482

ABSTRACT

Multidrug resistance protein 1 (MRP1) is an ATP-binding cassette transporter that confers multidrug resistance on tumor cells. Much convincing evidence has accumulated that MRP1 transports most substances in a GSH-dependent manner. On the other hand, several reports have revealed that MRP1 can transport some substrates independently of GSH; however, the importance of GSH-independent transport activity is not well established and the mechanistic differences between GSH-dependent and -independent transport by MRP1 are unclear. We previously demonstrated that the amino acids W261 and K267 in the L0 region of MRP1 were important for leukotriene C4 (LTC4) transport activity of MRP1 and for GSH-dependent photolabeling of MRP1 with azidophenyl agosterol-A (azidoAG-A). In this paper, we further tested the effect of W222L, W223L and R230A mutations in MRP1, designated dmL0MRP1, on MRP1 transport activity. SN-38 is an active metabolic form of CPT-11 that is one of the most promising anti-cancer drugs. Membrane vesicles prepared from cells expressing dmL0MRP1 could transport SN-38, but not LTC4 or estradiol-17 (beta-D-glucuronate), and could not be photolabeled with azidoAG-A. These data suggested that SN-38 was transported by a different mechanism than that of GSH-dependent transport. Understanding the GSH-independent transport mechanism of MRP1, and identification of drugs that are transported by this mechanism, will be critical for combating MRP1-mediated drug resistance. We performed a pairwise comparison of compounds that are transported by MRP1 in a GSH-dependent or -independent manner. These data indicated that it may be possible to predict compounds that are transported by MRP1 in a GSH-independent manner.


Subject(s)
Camptothecin/analogs & derivatives , Estradiol/pharmacokinetics , Leukotriene C4/pharmacokinetics , Multidrug Resistance-Associated Proteins/physiology , Base Sequence , Camptothecin/pharmacokinetics , Cell Line , Chromatography, High Pressure Liquid , DNA Primers , Humans , Irinotecan , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Photoaffinity Labels
15.
Mol Cancer Ther ; 1(14): 1327-34, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12516966

ABSTRACT

Ecteinascidin 743 (Et-743) is a novel anticancer agent forming covalent guanine adducts at specific sites in the DNA minor groove. Et-743 has a unique mechanism of action because it kills cancer cells by poisoning transcription-coupled nucleotide excision repair. Recent studies suggested a complex relationship between P-glycoprotein (P-gp)/MDR1 and Et-743. On one hand, Et-743 was reported to down-regulate the MDR1 promoter in vitro. On the other hand, P-gp overexpression was hypothesized to contribute to Et-743 resistance in an ovarian cell line. The present study was performed to further investigate the relationship between P-gp/MDR1 and the activity of Et-743. First, we found no P-gp/MDR1 overexpression (mRNA and protein levels) in two independently generated Et-743-resistant human colon carcinoma cell lines (HCT116/ER5 and SW480/ER0.5). Secondly, we found no cross-resistance to Et-743 in two well-characterized P-gp/MDR1-overexpressing cell lines (KB-8-5 and KB-C-2). Third, Et-743 pretreatment enhanced the cytotoxicity and the cellular accumulation of doxorubicin and vincristine in P-gp/MDR1-overexpressing KB-8-5/KB-C-2 cell lines. Fourth, we observed P-gp/MDR1 down-regulation by Et-743 in KB-C-2 cells. These results indicate that Et-743 does not select for the emergence of a P-gp phenotype in all cell lines made resistant to Et-743 and that P-gp overexpression is not sufficient to confer resistance to Et-743. Furthermore, Et-743 is an effective agent in P-gp-overexpressing cells. Et-743 can potentiate the activity of other chemotherapeutic agents by down-regulating P-gp/MDR1, suggesting that the combination of Et-743 and chemotherapeutic agents that are substrates for P-gp/MDR1 may be valuable in the clinic.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Dioxoles/pharmacology , Drug Resistance, Neoplasm , Isoquinolines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival , Coloring Agents/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Doxorubicin/pharmacology , Electrophoresis, Polyacrylamide Gel , Humans , Immunoblotting , Inhibitory Concentration 50 , Models, Chemical , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tetrahydroisoquinolines , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Trabectedin , Tumor Cells, Cultured , Vincristine/pharmacology
16.
Br J Pharmacol ; 138(8): 1553-61, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12721111

ABSTRACT

1. Human multidrug resistance protein 1 (MRP1) is a 190 kDa membrane glycoprotein that confers multidrug resistance (MDR) to tumor cells. We recently demonstrated that glutathione (GSH) is required for the labelling of the C-terminal half of MRP1 with a photoanalog of agosterol A (azido AG-A). In this study, we further characterized the GSH-dependent photolabelling site of azido AG-A on MRP1. 2. An epitope-inserted MRP1, MRP1 1222HA, which has two hemagglutinin A (HA) epitopes in the extracellular loop between transmembrane segment (TM) 16 and TM17 of the transporter, could bind azido AG-A in a GSH-dependent manner. 3. Protease digestion of the photolabelled MRP1 1222HA, followed by immunoprecipitation with an anti-HA antibody suggested that the GSH-dependent azido AG-A photolabelling site on MRP1 resides in the region within TM14-17 and the cytoplasmic region proximate to the C-terminus of TM17. 4. Arg(1210) in human MRP2 that corresponds to Arg(1202) in human MRP1 has an important role in the transporting activity of MRP2. Therefore, we replaced the Arg residue at position 1202 of MRP1 with Gly. Whereas photolabelling of the mutant MRP1 R1202G was greatly reduced, it retained leukotriene C(4) (LTC(4)) transport activity and conferred Vincristine resistance in LLC-PK1 cells. 5. In summary, this study demonstrated that the GSH-dependent azido AG-A photolabelling site on MRP1 resides in the region within TM14-17 and the cytoplasmic region proximate to the C-terminus of TM17. The charged amino acid Arg(1202) proximate to TM helix 16 is of critical importance for the GSH-dependent photolabelling of MRP1 with azido AG-A. Arg(1202) itself or the region nearby Arg(1202) may be involved in azido AG-A photolabelling.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Glutathione/analysis , Photoaffinity Labels/analysis , Sterols/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Animals , Cell Line , Immunochemistry , Insecta , Multidrug Resistance-Associated Protein 2 , Sterols/chemistry , Swine
17.
Cancer Lett ; 187(1-2): 111-9, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12359358

ABSTRACT

A newly synthesized 1,4-benzothiazipine derivate, 4-[3-(4-benzylpiperidin-1-yl) propionyl]-7-methoxy-2,3,4,5-tetrahydro-1, 4-benzothiazepine monohydrochloride (JTV-519) was examined for its ability to reverse P-glycoprotein (P-gp) and multidrug resistance protein 1 (MRP1) mediated multidrug resistance (MDR) in K562/MDR and KB/MRP cells, respectively. JTV-519 at 3 microM reversed the resistance of K562/MDR cells to vincristine (VCR), taxol, etoposide (VP16), adriamycin (ADM) and actinomycin D and at 0.5 or 1 microM reversed their resistance to STI571. JTV-519 at 10 microM enhanced the accumulation of ADM in K562/MDR cells to the level in parental K562 cells and inhibited the efflux of ADM from K562/MDR cells. Photoaffinity labeling of P-gp with 3H-azidopine was almost completely inhibited by 500 microM JTV-519. JTV-519 at 3 microM also partially reversed the resistance of KB/MRP cells to VCR and at 500 microM partially inhibited the photoaffinity labeling of MRP1 with (125)I-II-azidophenyl agosterol A (125I-azidoAG-A). These results suggest that JTV-519 reversed the resistance to the anti-cancer agents in P-gp and MRP1 overexpressing multidrug-resistant cells by directly binding to P-gp and MRP1, and competitively inhibiting transport of the anti-cancer agents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Thiazepines/pharmacology , Antineoplastic Agents/pharmacology , Benzamides , Cell Membrane , Cell Survival/drug effects , Doxorubicin/pharmacology , Flow Cytometry , Humans , Imatinib Mesylate , Immunoblotting , Multidrug Resistance-Associated Proteins/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
18.
Neurosci Lett ; 345(1): 5-8, 2003 Jul 10.
Article in English | MEDLINE | ID: mdl-12809975

ABSTRACT

In this study, we investigated the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) gastric administration on the expression of neuronal nitric oxide synthase (nNOS) and the NADPH-diaphorase (NADPH-d) activities in the brain of the Long-Evans rat. A single dose of TCDD (dissolved in olive oil, 50 microg/kg) or olive oil alone was administered to the rats by gavage. nNOS Western blotting experiment indicated a marked decrease in nNOS immunoreactivity at 1 and 2 weeks after TCDD treatment. NADPH-d histochemistry results showed a marked decrease in the number of NADPH-d stained cell bodies in the paraventricular hypothalamic nucleus, lateral hypothalamic area and perifornical nucleus in the TCDD-treated rats. The present study suggests that TCDD administration down-regulates nitric oxide product in the hypothalamus, which may be partially responsible for TCDD-induced feeding inhibition.


Subject(s)
Down-Regulation , Environmental Pollutants/adverse effects , Hypothalamus/drug effects , NADPH Dehydrogenase/biosynthesis , Nitric Oxide Synthase/biosynthesis , Polychlorinated Dibenzodioxins/adverse effects , Animals , Hypothalamus/metabolism , Immunohistochemistry , Male , Nitric Oxide Synthase Type I , Rats , Rats, Long-Evans
19.
Oncol Res ; 14(1): 39-48, 2003.
Article in English | MEDLINE | ID: mdl-14552590

ABSTRACT

Agosterol A (AG-A) is a novel agent that reverses P-glycoprotein (P-gp) and multidrug resistance protein-1 (MRP1)-meditated multidrug resistance (MDR). We have synthesized [125I]11-azidophenyl agosterol A (azidoAG-A), a photoaffinity analog of AG-A, and characterized its binding to P-gp in membrane vesicles prepared from multidrug-resistant P-gp-overexpressing KB-C2 cells. The photoanalog photolabeled intact P-gp and both the N- and C-terminal fragments of P-gp. [125I]AzidoAG-A is transported by P-gp and the intracellular accumulation of both [125I]azidoAG-A and [3H]AG-A in KB-C2 cells was lower than that in the parental drug-sensitive KB-3-1 cells. [125I]AzidoAG-A bound to the drug binding site(s) on P-gp because photoaffinity labeling of P-gp was inhibited by a variety of known P-gp substrates, including anticancer, reversing, and anti-human immunodeficiency virus (HIV) agents. The binding of [125I]azidoAG-A to P-gp differs from the binding of other photolabeled probes such as iodoaryl-azidoprazosin (IAAP) to P-gp and from the binding of [125I]azidoAG-A to MRP1 based on the differing effects of flupentixol and glutathione (GSH) on their binding. Thus, [125I]azidoAG-A will be a useful tool to elucidate the structure and function of P-gp because it directly binds to the drug binding site(s) on P-gp, is transported by P-gp, and exhibits different P-gp binding characteristics than IAAP.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinoma, Squamous Cell/metabolism , Photoaffinity Labels , Sterols/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Membrane/metabolism , Drug Resistance, Multiple , Humans , Precipitin Tests , Sterols/chemistry
20.
J Biol Chem ; 284(35): 23251-9, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19567877

ABSTRACT

The mechanisms involved in the targeting of neuronal nicotinic acetylcholine receptors (AChRs), critical for their functional organization at neuronal synapses, are not well understood. We have identified a novel functional association between alpha4beta2 AChRs and the presynaptic cell adhesion molecule, neurexin-1beta. In non-neuronal tsA 201 cells, recombinant neurexin-1beta and mature alpha4beta2 AChRs form complexes. alpha4beta2 AChRs and neurexin-1beta also coimmunoprecipitate from rat brain lysates. When exogenous alpha4beta2 AChRs and neurexin-1beta are coexpressed in hippocampal neurons, they are robustly targeted to hemi-synapses formed between these neurons and cocultured tsA 201 cells expressing neuroligin-1, a postsynaptic binding partner of neurexin-1beta. The extent of synaptic targeting is significantly reduced in similar experiments using a mutant neurexin-1beta lacking the extracellular domain. Additionally, when alpha4beta2 AChRs, alpha7 AChRs, and neurexin-1beta are coexpressed in the same neuron, only the alpha4beta2 AChR colocalizes with neurexin-1beta at presynaptic terminals. Collectively, these data suggest that neurexin-1beta targets alpha4beta2 AChRs to presynaptic terminals, which mature by trans-synaptic interactions between neurexins and neuroligins. Interestingly, human neurexin-1 gene dysfunctions have been implicated in nicotine dependence and in autism spectrum disorders. Our results provide novel insights as to possible mechanisms by which dysfunctional neurexins, through downstream effects on alpha4beta2 AChRs, may contribute to the etiology of these neurological disorders.


Subject(s)
Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Receptors, Nicotinic/metabolism , Animals , Cells, Cultured , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Binding , Protein Transport , Rats , Receptors, Nicotinic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL