Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Oncogene ; 43(22): 1655-1668, 2024 May.
Article in English | MEDLINE | ID: mdl-38658776

ABSTRACT

Prostate cancer (CaP) remains the second leading cause of cancer deaths in western men. CaP mortality results from diverse molecular mechanisms that mediate resistance to the standard of care treatments for metastatic disease. Recently, alternative splicing has been recognized as a hallmark of CaP aggressiveness. Alternative splicing events cause treatment resistance and aggressive CaP behavior and are determinants of the emergence of the two major types of late-stage treatment-resistant CaP, namely castration-resistant CaP (CRPC) and neuroendocrine CaP (NEPC). Here, we review recent multi-omics data that are uncovering the complicated landscape of alternative splicing events during CaP progression and the impact that different gene transcript isoforms can have on CaP cell biology and behavior. We discuss renewed insights in the molecular machinery by which alternative splicing occurs and contributes to the failure of systemic CaP therapies. The potential for alternative splicing events to serve as diagnostic markers and/or therapeutic targets is explored. We conclude by considering current challenges and promises associated with splicing-modulating therapies, and their potential for clinical translation into CaP patient care.


Subject(s)
Alternative Splicing , Disease Progression , Drug Resistance, Neoplasm , Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Humans , Alternative Splicing/genetics , Male , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Animals
2.
Genomics ; 116(1): 110772, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38158140

ABSTRACT

Identifying biomarkers for diagnosing Major Depressive Disorder (MDD), assessing its severity, and guiding treatment is crucial. We conducted whole genome transcriptomic study in North Indian population, and analyzed biochemical parameters. Our longitudinal study investigated gene-expression profiles from 72 drug-free MDD patients and 50 healthy controls(HCs) at baseline and 24 patients after 12-weeks of treatment. Gene expression analyses identified differentially expressed genes(DEGs) associated with MDD susceptibility, symptom severity and treatment response, independently validated by qPCR. Hierarchical clustering revealed distinct expression patterns between MDD and HCs, also between mild and severe cases. Enrichment analyses of significant DEGs revealed inflammatory, apoptosis, and immune-related pathways in MDD susceptibility, severity, and treatment response. Simultaneously, we assessed thirty biochemical parameters in the same cohort, showed significant differences between MDD and HCs in 13 parameters with monocytes, eosinophils, creatinine, SGPT, and total protein remained independent predictors of MDD in a multivariate-regression model. Our study supports the role of altered immune/inflammatory signaling in MDD pathophysiology, offering clinically relevant biochemical parameters and insights into transcriptomic gene regulation in MDD pathogenesis and treatment response.


Subject(s)
Depressive Disorder, Major , Humans , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Longitudinal Studies , Antidepressive Agents/therapeutic use , Gene Expression Profiling , Transcriptome
3.
Cancer Res ; 83(24): 4142-4160, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37801613

ABSTRACT

Prostate cancer remains the second leading cause of cancer death in men in Western cultures. A deeper understanding of the mechanisms by which prostate cancer cells divide to support tumor growth could help devise strategies to overcome treatment resistance and improve survival. Here, we identified that the mitotic AGC family protein kinase citron kinase (CIT) is a pivotal regulator of prostate cancer growth that mediates prostate cancer cell interphase progression. Increased CIT expression correlated with prostate cancer growth induction and aggressive prostate cancer progression, and CIT was overexpressed in prostate cancer compared with benign prostate tissue. CIT overexpression was controlled by an E2F2-Skp2-p27 signaling axis and conferred resistance to androgen-targeted treatment strategies. The effects of CIT relied entirely on its kinase activity. Conversely, CIT silencing inhibited the growth of cell lines and xenografts representing different stages of prostate cancer progression and treatment resistance but did not affect benign epithelial prostate cells or nonprostatic normal cells, indicating a potential therapeutic window for CIT inhibition. CIT kinase activity was identified as druggable and was potently inhibited by the multikinase inhibitor OTS-167, which decreased the proliferation of treatment-resistant prostate cancer cells and patient-derived organoids. Isolation of the in vivo CIT substrates identified proteins involved in diverse cellular functions ranging from proliferation to alternative splicing events that are enriched in treatment-resistant prostate cancer. These findings provide insights into the regulation of aggressive prostate cancer cell behavior by CIT and identify CIT as a functionally diverse and druggable driver of prostate cancer progression. SIGNIFICANCE: The poorly characterized protein kinase citron kinase is a therapeutic target in prostate cancer that drives tumor growth by regulating diverse substrates, which control several hallmarks of aggressive prostate cancer progression. See related commentary by Mishra et al., p. 4008.


Subject(s)
Prostate , Prostatic Neoplasms , Protein Kinases , Humans , Male , Cell Line, Tumor , Cell Proliferation , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Kinases/metabolism , Signal Transduction
4.
Mol Pharmacol ; 103(3): 145-157, 2023 03.
Article in English | MEDLINE | ID: mdl-36414374

ABSTRACT

Despite the progress made in the development of new antiepileptic drugs (AEDs), poor response to them is a rising concern in epilepsy treatment. Of several hypotheses explaining AED treatment failure, the most promising theory is the overexpression of multidrug transporters belonging to ATP-binding cassette (ABC) transporter family at blood-brain barrier. Previous data show that AEDs themselves can induce these transporters, in turn affecting their own brain bioavailability. Presently, this induction and the underlying regulatory mechanism involved at human blood-brain barrier is not well elucidated. Herein, we sought to explore the effect of most prescribed first- and second-line AEDs on multidrug transporters in human cerebral microvascular endothelial cells, hCMEC/D3. Our work demonstrated that exposure of these cells to valproic acid (VPA) induced mRNA, protein, and functional activity of breast cancer resistance protein (BCRP/ABCG2). On examining the substrate interaction status of AEDs with BCRP, VPA, phenytoin, and lamotrigine were found to be potential BCRP substrates. Furthermore, we observed that siRNA-mediated knockdown of peroxisome proliferator-activated receptor alpha (PPARα) or use of PPARα antagonist, resulted in attenuation of VPA-induced BCRP expression and transporter activity. VPA was found to increase PPARα expression and trigger its translocation from cytoplasm to nucleus. Findings from chromatin immunoprecipitation and luciferase assays showed that VPA enhances the binding of PPARα to its response element in the ABCG2 promoter, resulting in elevated ABCG2 transcriptional activity. Taken together, these in vitro findings highlight PPARα as the potential molecular target to prevent VPA-mediated BCRP induction, which may have important implications in VPA pharmacoresistance. SIGNIFICANCE STATEMENT: Induction of multidrug transporters at blood-brain barrier can largely affect the bioavailability of the substrate antiepileptic drugs in the brains of patients with epilepsy, thus affecting their therapeutic efficacy. The present study reports a mechanistic pathway of breast cancer resistance protein (BCRP/ABCG2) upregulation by valproic acid in human brain endothelial cells via peroxisome proliferator-activated receptor alpha involvement, thereby providing a potential strategy to prevent valproic acid pharmacoresistance in epilepsy.


Subject(s)
Breast Neoplasms , Epilepsy , Humans , Female , PPAR alpha/metabolism , Valproic Acid/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Anticonvulsants/pharmacology , Up-Regulation , Endothelial Cells/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Brain/metabolism , Membrane Transport Proteins/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Epilepsy/drug therapy , Epilepsy/metabolism , Breast Neoplasms/metabolism
5.
Front Genet ; 12: 722221, 2021.
Article in English | MEDLINE | ID: mdl-34956307

ABSTRACT

Previously, we demonstrated an integrated genomic convergence and network analysis approach to identify the candidate genes associated with the complex neurodegenerative disorder, Alzheimer's disease (AD). Here, we performed a pilot study to validate the in silico approach by studying the association of genetic variants from three identified critical genes, APOE, EGFR, and ACTB, with AD. A total of 103 patients with AD and 146 healthy controls were recruited. A total of 46 single-nucleotide polymorphisms (SNPs) spanning the three genes were genotyped, of which only 19 SNPs were included in the final analyses after excluding non-polymorphic and Hardy-Weinberg equilibrium-violating SNPs. Apart from our previously reported APOE ε4, four other SNPs in APOE (rs405509, rs7259620, -rs769449, and rs7256173), one in EGFR (rs6970262), and one in ACTB (rs852423) showed a significant association with AD (p < 0.05). Our results validate the reliability of genomic convergence and network analysis approach in identifying the AD-associated candidate genes.

6.
Cell Mol Life Sci ; 78(21-22): 6887-6939, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34586444

ABSTRACT

The adenosine triphosphate (ATP)-binding cassette efflux transporter G2 (ABCG2) was originally discovered in a multidrug-resistant breast cancer cell line. Studies in the past have expanded the understanding of its role in physiology, disease pathology and drug resistance. With a widely distributed expression across different cell types, ABCG2 plays a central role in ATP-dependent efflux of a vast range of endogenous and exogenous molecules, thereby maintaining cellular homeostasis and providing tissue protection against xenobiotic insults. However, ABCG2 expression is subjected to alterations under various pathophysiological conditions such as inflammation, infection, tissue injury, disease pathology and in response to xenobiotics and endobiotics. These changes may interfere with the bioavailability of therapeutic substrate drugs conferring drug resistance and in certain cases worsen the pathophysiological state aggravating its severity. Considering the crucial role of ABCG2 in normal physiology, therapeutic interventions directly targeting the transporter function may produce serious side effects. Therefore, modulation of transporter regulation instead of inhibiting the transporter itself will allow subtle changes in ABCG2 activity. This requires a thorough comprehension of diverse factors and complex signaling pathways (Kinases, Wnt/ß-catenin, Sonic hedgehog) operating at multiple regulatory levels dictating ABCG2 expression and activity. This review features a background on the physiological role of transporter, factors that modulate ABCG2 levels and highlights various signaling pathways, molecular mechanisms and genetic polymorphisms in ABCG2 regulation. This understanding will aid in identifying potential molecular targets for therapeutic interventions to overcome ABCG2-mediated multidrug resistance (MDR) and to manage ABCG2-related pathophysiology.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Animals , Humans , Neoplasm Proteins/genetics , Polymorphism, Genetic/genetics , Signal Transduction/genetics
7.
Int J Mol Sci ; 21(20)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096746

ABSTRACT

Epilepsy, a neurological disease characterized by recurrent seizures, is highly heterogeneous in nature. Based on the prevalence, epilepsy is classified into two types: common and rare epilepsies. Common epilepsies affecting nearly 95% people with epilepsy, comprise generalized epilepsy which encompass idiopathic generalized epilepsy like childhood absence epilepsy, juvenile myoclonic epilepsy, juvenile absence epilepsy and epilepsy with generalized tonic-clonic seizure on awakening and focal epilepsy like temporal lobe epilepsy and cryptogenic focal epilepsy. In 70% of the epilepsy cases, genetic factors are responsible either as single genetic variant in rare epilepsies or multiple genetic variants acting along with different environmental factors as in common epilepsies. Genetic testing and precision treatment have been developed for a few rare epilepsies and is lacking for common epilepsies due to their complex nature of inheritance. Precision medicine for common epilepsies require a panoramic approach that incorporates polygenic background and other non-genetic factors like microbiome, diet, age at disease onset, optimal time for treatment and other lifestyle factors which influence seizure threshold. This review aims to comprehensively present a state-of-art review of all the genes and their genetic variants that are associated with all common epilepsy subtypes. It also encompasses the basis of these genes in the epileptogenesis. Here, we discussed the current status of the common epilepsy genetics and address the clinical application so far on evidence-based markers in prognosis, diagnosis, and treatment management. In addition, we assessed the diagnostic predictability of a few genetic markers used for disease risk prediction in individuals. A combination of deeper endo-phenotyping including pharmaco-response data, electro-clinical imaging, and other clinical measurements along with genetics may be used to diagnose common epilepsies and this marks a step ahead in precision medicine in common epilepsies management.


Subject(s)
Epilepsy/drug therapy , Epilepsy/genetics , DNA Copy Number Variations , Epilepsy/diagnosis , Epilepsy, Absence/genetics , Epilepsy, Generalized/genetics , Genetic Markers , Humans , Pharmacogenomic Testing , Precision Medicine/methods , Prognosis , Seizures/genetics , Time Factors
8.
Sci Rep ; 10(1): 2546, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054883

ABSTRACT

Antiepileptic drug therapy has significant inter-patient variability in response towards it. The current study aims to understand this variability at the molecular level using microarray-based analysis of peripheral blood gene expression profiles of patients receiving valproate (VA) monotherapy. Only 10 unique genes were found to be differentially expressed in VA responders (n = 15) and 6 genes in the non-responders (n = 8) (fold-change >2, p < 0.05). PTGS2 which encodes cyclooxygenase-2, COX-2, showed downregulation in the responders compared to the non-responders. PTGS2/COX-2 mRNA profiles in the two groups corresponded to their plasma profiles of the COX-2 product, prostaglandin E2 (PGE2). Since COX-2 is believed to regulate P-glycoprotein (P-gp), a multidrug efflux transporter over-expressed at the blood-brain barrier (BBB) in drug-resistant epilepsy, the pathway connecting COX-2 and P-gp was further explored in vitro. Investigation of the effect of VA upon the brain endothelial cells (hCMEC/D3) in hyperexcitatory conditions confirmed suppression of COX-2-dependent P-gp upregulation by VA. Our findings suggest that COX-2 downregulation by VA may suppress seizure-mediated P-gp upregulation at the BBB leading to enhanced drug delivery to the brain in the responders. Our work provides insight into the association of peripheral PTGS2/COX-2 expression with VA efficacy and the role of COX-2 as a potential therapeutic target for developing efficacious antiepileptic treatment.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cyclooxygenase 2/genetics , Epilepsy/drug therapy , Valproic Acid/administration & dosage , Adult , Blood-Brain Barrier/drug effects , Endothelial Cells/drug effects , Epilepsy/genetics , Epilepsy/pathology , Female , Gene Expression Regulation/drug effects , Humans , Male , Transcriptome/drug effects , Transcriptome/genetics
9.
Article in English | MEDLINE | ID: mdl-32007745

ABSTRACT

Prostaglandin E2 (PGE2), a physiologically active lipid compound, is increased in several diseases characterized by chronic inflammation. To determine its significance in epilepsy-associated inflammation and response to antiepileptic drug (AED), we evaluated the plasma PGE2 (median, pg/ml) levels in drug-free patients with epilepsy (N = 34) and patients receiving AED monotherapy (N = 55) in addition to that in healthy controls (N = 34). When compared to controls, plasma PGE2 levels were significantly elevated in all drug-free patients independent of the type of epilepsy (137.2 versus 475.7 pg/ml, p < 0.0001). Among the patients receiving AED monotherapy, only valproate responders showed a significant decrease compared to both drug-free patients (232.1 versus 475.7 pg/ml, p < 0.01) as well as valproate non-responders (232.1 versus 611.9 pg/ml, p < 0.0001). Both responders and non-responders on phenytoin or carbamazepine monotherapy had elevated PGE2 levels similar to drug-free patients. In addition, no difference was observed in plasma profiles of PGE2 precursor, arachidonic acid among the groups. Our work presents the clinical evidence of the association between plasma PGE2 levels and valproate efficacy in patients with epilepsy.


Subject(s)
Anticonvulsants/administration & dosage , Epilepsy/drug therapy , Prostaglandins E/blood , Adolescent , Adult , Anticonvulsants/pharmacology , Carbamazepine/administration & dosage , Carbamazepine/pharmacology , Case-Control Studies , Epilepsy/metabolism , Female , Humans , Male , Phenytoin/administration & dosage , Phenytoin/pharmacology , Treatment Outcome , Valproic Acid/administration & dosage , Valproic Acid/pharmacology , Young Adult
10.
Genomics ; 112(1): 218-224, 2020 01.
Article in English | MEDLINE | ID: mdl-30826443

ABSTRACT

Heterogeneity in epilepsy often interferes with its diagnosis as well as treatment. To examine this heterogeneity at transcriptomic level, we performed whole-genome mRNA expression profiling in whole blood samples from 34 patients with epilepsy (PWE) (idiopathic, n = 13; cryptogenic, n = 9; and symptomatic, n = 12) and 41 healthy controls (HC) using Illumina HT-12 Expression Beadchip v4 microarray. In silico analysis using R software identified 165 genes to be significantly differentially expressed in PWE compared to HC (fold change>1.3, p < 0.05). Hierarchical clustering of resultant DEGs segregated idiopathic epilepsy from the rest of the epilepsy classes as well as HC. The class also displayed the most differential expression pattern with the highest number of DEGs among the three epilepsy classes. Gene ontology analysis revealed several biologically relevant inflammatory and other immune-related pathways. Our study provides insight into the relevance of altered blood gene expression patterns in understanding epilepsy and its etiologic classes.


Subject(s)
Epilepsy/genetics , Transcriptome , Adolescent , Adult , Cluster Analysis , Epilepsy/blood , Epilepsy/classification , Female , Gene Expression Profiling , Gene Ontology , Humans , Male , Real-Time Polymerase Chain Reaction , Young Adult
11.
J Neuroinflammation ; 16(1): 197, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31666079

ABSTRACT

Epilepsy, a common multifactorial neurological disease, affects about 69 million people worldwide constituting nearly 1% of the world population. Despite decades of extensive research on understanding its underlying mechanism and developing the pharmacological treatment, very little is known about the biological alterations leading to epileptogenesis. Due to this gap, the currently available antiepileptic drug therapy is symptomatic in nature and is ineffective in 30% of the cases. Mounting evidences revealed the pathophysiological role of neuroinflammation in epilepsy which has shifted the focus of epilepsy researchers towards the development of neuroinflammation-targeted therapeutics for epilepsy management. Markedly increased expression of key inflammatory mediators in the brain and blood-brain barrier may affect neuronal function and excitability and thus may increase seizure susceptibility in preclinical and clinical settings. Cyclooxygenase-2 (COX-2), an enzyme synthesizing the proinflammatory mediators, prostaglandins, has widely been reported to be induced during seizures and is considered to be a potential neurotherapeutic target for epilepsy management. However, the efficacy of such therapy involving COX-2 inhibition depends on various factors viz., therapeutic dose, time of administration, treatment duration, and selectivity of COX-2 inhibitors. This article reviews the preclinical and clinical evidences supporting the role of COX-2 in seizure-associated neuroinflammation in epilepsy and the potential clinical use of COX-2 inhibitors as a future strategy for epilepsy treatment.


Subject(s)
Anticonvulsants/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Management , Epilepsy/diagnosis , Epilepsy/drug therapy , Animals , Clinical Trials as Topic/methods , Epilepsy/enzymology , Forecasting , Humans
12.
Neurol India ; 66(4): 1052-1059, 2018.
Article in English | MEDLINE | ID: mdl-30038093

ABSTRACT

BACKGROUND: Nearly 40%-50% of the individuals fail to respond to first line antiepileptic drug (AED) monotherapy and 30% are refractory, which calls for the need to recognize predictive markers for treatment failure. This study aims to identify clinical factors predictive of a poor prognosis in patients on AED therapy. MATERIALS AND METHODS: A prospective follow-up study involving 1056 patients with epilepsy (PWE) aged 5-67 years from North India on phenytoin (PHT, n = 247), carbamazepine (CBZ, n = 369), valproate (VA, n = 271), phenobarbital (PB, n = 50), and multitherapy (MultiT, n = 119) was conducted between 2005 and 2015. Seizure and epilepsy types were diagnosed based on the classifications by the International League Against Epilepsy (ILAE). Patients remaining seizure-free during the past 1 year were assigned to the "no seizure" group and patients experiencing seizure recurrence were assigned to the "recurrent seizures" group. RESULTS: Of the total, 786 (74.4%) patients were successfully followed up with 60% achieving 1-year seizure remission. Seizure recurrence was observed in the remaining 40% of the patients with a high likelihood in patients with the disease onset at ≤5 years of age [55% vs. 38%, P = 0.0016, odds ratio (OR) = 2.02 (95% confidence interval (CI) = 1.31-3.13)], in patients with cryptogenic epilepsy than with idiopathic/symptomatic epilepsy (48% vs. 32%, P = 0.0049, OR = 1.61 [95% CI = 1.16-2.24]), and in patients with pretreatment seizure frequency ≥12/year (46% vs. 27%, P < 0.0001, OR = 2.21 [95% CI = 1.61-3.05]). Logistic regression analysis also revealed a significant association of seizure recurrence (P < 0.05) with the three variables. CONCLUSION: Our findings suggest that an early disease onset, cryptogenic epilepsy, and a higher pretreatment seizure frequency are related to a poor prognosis or poor remission in people with epilepsy (PWE) on AED therapy.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Treatment Outcome , Adolescent , Adult , Aged , Asian People , Child , Child, Preschool , Female , Humans , India , Male , Middle Aged , Prognosis , Prospective Studies , Seizures/prevention & control , Young Adult
13.
BMC Med Genomics ; 10(1): 56, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28927418

ABSTRACT

BACKGROUND: PD is a progressive neurodegenerative disorder commonly treated by levodopa. The findings from genetic studies on adverse effects (ADRs) and levodopa efficacy are mostly inconclusive. Here, we aim to identify predictive genetic biomarkers for levodopa response (LR) and determine common molecular link with disease susceptibility. A systematic review for LR was conducted for ADR, and drug efficacy, independently. All included articles were assessed for methodological quality on 14 parameters. GWAS of PD were also reviewed. Protein-protein interaction (PPI) analysis using STRING and functional enrichment using WebGestalt was performed to explore the common link between LR and PD. RESULTS: From 37 candidate studies on levodopa toxicity, 18 genes were found associated, of which, CAn STR 13, 14 (DRD2) was most significantly associated with dyskinesia, followed by rs1801133 (MTHFR) with hyper-homocysteinemia, and rs474559 (HOMER1) with hallucination. Similarly, 8 studies on efficacy resulted in 4 genes in which rs28363170, rs3836790 (SLC6A3) and rs4680 (COMT), were significant. To establish the molecular connection between LR with PD, we identified 35 genes significantly associated with PD. With 19 proteins associated with LR and 35 with PD, two independent PPI networks were constructed. Among the 67 nodes (263 edges) in LR, and 62 nodes (190 edges) in PD pathophysiology, UBC, SNCA, FYN, SRC, CAMK2A, and SLC6A3 were identified as common potential candidates. CONCLUSION: Our study revealed the genetically significant polymorphism concerning the ADRs and levodopa efficacy. The six common genes may be used as predictive markers for therapy optimization and as putative drug target candidates.


Subject(s)
Levodopa/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Genetic Predisposition to Disease , Humans , Levodopa/adverse effects , Levodopa/therapeutic use , Parkinson Disease/metabolism , Protein Interaction Maps
14.
Pharm Res ; 34(7): 1444-1458, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28432535

ABSTRACT

PURPOSE: Over expression of ATP-binding cassette transporters is considered one of the major reasons for non-responsiveness to antiepileptic drugs. Carbamazepine (CBZ), one of first line antiepileptic drug is known to influence ABCC2 expression but its exact molecular mechanism is unknown. METHODS: We investigated the effect of CBZ on expression of ABCC2 and pregnane X receptor (PXR) in HepG2 cell line and compared with hyperforin (agonist of PXR) and ketoconazole (antagonist of PXR) through realtime PCR and western blot assay. Involvement of PXR was demonstrated through nuclear translocation and RNA interference and related effect of CBZ on ABCC2 through functional activity assay. Molecular docking and dynamic simulation approach was used to understand the interaction of CBZ with PXR. RESULTS: CBZ and hyperforin increased the PXR and ABCC2 expression whereas reversed when present it in combination with ketoconazole. Experiments confirmed CBZ induced ABCC2 expression is PXR dependent. Molecular dynamic (MD) simulation and in vitro experiment indicated possibility of CBZ to be PXR agonist and PXR residue Gln285 to be important for CBZ-PXR interaction. CONCLUSIONS: CBZ alters the functional activity of ABCC2 through PXR, which in turn can interfere with therapy. Mutational analysis of residues revealed the importance of Gln285 in ligand interaction.


Subject(s)
Anticonvulsants/chemistry , Carbamazepine/chemistry , Multidrug Resistance-Associated Proteins/chemistry , Receptors, Steroid/chemistry , Active Transport, Cell Nucleus , Anticonvulsants/metabolism , Anticonvulsants/pharmacology , Binding, Competitive , Carbamazepine/pharmacology , Cell Nucleus/metabolism , Computer Simulation , Hep G2 Cells , Humans , Ketoconazole/chemistry , Ketoconazole/pharmacology , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Mutation , Phloroglucinol/analogs & derivatives , Phloroglucinol/chemistry , Phloroglucinol/pharmacology , Pregnane X Receptor , Protein Binding , RNA Interference , Receptors, Steroid/agonists , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/genetics , Terpenes/chemistry , Terpenes/pharmacology
15.
CNS Neurosci Ther ; 22(9): 740-57, 2016 09.
Article in English | MEDLINE | ID: mdl-27245092

ABSTRACT

AIM: The present study aimed to evaluate association of genetic variants on drug response and therapy optimization parameters in patients treated with first-line antiepileptic drugs (AEDs). Genetic variants from ion channels, their functionally related genes, and synaptic vesicle cycle (SVC) genes with a potential role in epilepsy pathophysiology were thus prioritized. METHODS: A total of 12 genes from ion channels and related gene set and seven genes from SVC comprising 155 SNPs were genotyped and evaluated with drug response, dose levels, and drug levels in 408 patients with epilepsy. RESULTS: Both GABRA1 and SCN1A variants showed haplotypic and diplotypic associations in response to phenytoin (PHT). Diplotype analysis of GABRA1 variants revealed association of rs12658835|rs7735530 (AG/AG) (P-valuecorrected  = 0.034, OR = 3.75, 95% CI = 1.36-11.05) and rs12658835|rs7735530|rs7732641|rs2279020 (AGCA/AGCA) (P-valuecorrected  = 0.035, OR = 2.48, 95% CI = 0.96-6.41) with recurrent seizures. SCN1A haplotype rs6432860|rs3812718 (AC: P-valuecorrected  = 0.022, OR = 2.72, 95% CI = 1.39-5.35) and diplotype (AC/AC: P-valuecorrected  = 0.034, OR = 6.42, 95% CI = 1.10-65.76) were further observed to be associated with recurrent seizures. With respect to therapy optimization parameters, we observed significantly lower dose-adjusted drug levels at maximum dose of PHT in patients carrying AC/AC diplotype (P-value = 0.021). CONCLUSION: The results further substantiate the role of GABRA1 in PHT mode of action and contribution of SCN1A in response and therapy optimization with PHT monotherapy.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy , NAV1.1 Voltage-Gated Sodium Channel/genetics , Pharmacogenomic Variants/genetics , Phenytoin/therapeutic use , Polymorphism, Single Nucleotide/genetics , Receptors, GABA-A/genetics , Adolescent , Adult , Child , Epilepsy/blood , Epilepsy/drug therapy , Epilepsy/genetics , Female , Humans , India , Male , Young Adult
16.
Mol Neurobiol ; 53(7): 4833-64, 2016 09.
Article in English | MEDLINE | ID: mdl-26351077

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by loss of memory and other cognitive functions. AD can be classified into familial AD (FAD) and sporadic AD (SAD) based on heritability and into early onset AD (EOAD) and late onset AD (LOAD) based on age of onset. LOAD cases are more prevalent with genetically complex architecture. In spite of significant research focused on understanding the etiological mechanisms, search for diagnostic biomarker(s) and disease-modifying therapy is still on. In this article, we aim to comprehensively review AD literature on established etiological mechanisms including role of beta-amyloid and apolipoprotein E (APOE) along with promising newer etiological factors such as epigenetic modifications that have been associated with AD suggesting its multifactorial nature. As genomic studies have recently played a significant role in elucidating AD pathophysiology, a systematic review of findings from genome-wide linkage (GWL), genome-wide association (GWA), genome-wide expression (GWE), and epigenome-wide association studies (EWAS) was conducted. The availability of multi-dimensional genomic data has further coincided with the advent of computational and network biology approaches in recent years. Our review highlights the importance of integrative approaches involving genomics and systems biology perspective in elucidating AD pathophysiology. The promising newer approaches may provide reliable means of early and more specific diagnosis and help identify therapeutic interventions for LOAD.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Genomics/methods , Systems Biology/trends , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Animals , Apolipoproteins E/genetics , Epigenomics/methods , Epigenomics/trends , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Genome-Wide Association Study/trends , Genomics/trends , Humans , Systems Biology/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...