1
|
Majumder P, Hsu TI, Hu CJ, Huang JK, Lee YC, Hsieh YC, Ahsan A, Huang CC. Potential role of solid lipid curcumin particle (SLCP) as estrogen replacement therapy in mitigating TDP-43-related neuropathy in the mouse model of ALS disease. Exp Neurol 2024; 383:114999. [PMID: 39419433 DOI: 10.1016/j.expneurol.2024.114999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/10/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) was first identified in 1869, but it wasn't until the 2014 Ice Bucket Challenge that widespread attention was drawn to the disease. Since then, substantial research has been dedicated to developing treatments for ALS. Despite this, only three drugs - riluzole, edaravone and AMX0035, have been approved for clinical use, and they can only temporarily alleviate mild symptoms without significant disease modification or cure. Therefore, there remains a critical unmet need to identify disease modifying or curative therapies for ALS. The higher incidence and more severe progression of ALS and FTLD (frontotemporal lobar degeneration) observed in men and postmenopausal woman compared to young women suggests that sex hormones may significantly influence disease onset and progression. In both animal models and human clinical studies, 17β estradiol (E2) has been shown to delay and improve the outcomes of many neurodegenerative diseases. Here, we examined the role of TDP-43 in the regulation of estrogen-related enzymes, CYP19A1 and CYP3A4. In addition, we examined the impact of curcumin on the regulation of estrogen E2 levels and TDP-43-associated neuropathy as a potential therapeutic strategy for the treatment of FTLD and ALS. METHODS Prp-TDP-43A315T mice was used as a model of ALS/FTLD to examine the expression patterns of E2 and its biosynthesis and degradation enzymes, CYP19A1 and CYP3A4. Moreover, the molecular mechanisms and the potency of solid lipid curcumin particles (SLCP) as an E2 replacement therapy for TDP-43 associated neuropathy was analyzed. We further examined the survival rates and the pathological TDP43 patterns in female and male Prp-TDP-43A315T mice administrated with or without SLCP. In addition, the changed expression levels of enzymes corresponding to E2 biosynthesis and degradation in the spinal cord of female and male Prp-TDP-43A315T mice with or without SLCP were determined. RESULTS We found that in addition to E2, the expression patterns of CYP19A1 and CYP3A4 proteins differed between Prp-TDP-43A315T mice compared to wild-type control, suggesting that toxic phosphorylated TDP43 oligomers may disrupt the balance between CYP19A1 and CYP3A4 expression, leading to reduced estrogen biosynthesis and accelerated degradation. In addition, we found that oral administration of SLCP prolonged the survival rates in female Prp-TDP-43A315T mice and significantly reduced the pathological insoluble phosphorylated TDP-43 species. Furthermore, SLCP attenuated disease progression associated with TDP-43-related neuropathies through modulating estrogen biosynthesis and the activity of CYP450 enzymes. CONCLUSIONS Our results showed that Prp-TDP-43A315T mice exhibit altered estradiol levels. Moreover, we demonstrated the efficacy of SLCP as an estrogen replacement therapy in mitigating TDP-43-associated disease progression and pathogenesis. These findings suggest that SLCP could be a promising strategy to induce E2 expression for the treatment of ALS and FTLD.
Collapse
Affiliation(s)
- Pritha Majumder
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Chaur-Joug Hu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan; Neurology Department, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Yi-Chao Lee
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Chen Hsieh
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Asmar Ahsan
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan
| | - Chi-Chen Huang
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
2
|
Samriti, Kaur A, Kaur A, Goel RK. Ameliorative effect of diclofenac in rotenone corneal kindling model of drug-resistant epilepsy: Edge of dual COX and KMO inhibition. Brain Res 2024; 1846:149246. [PMID: 39304107 DOI: 10.1016/j.brainres.2024.149246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Epilepsy affects millions of people worldwide, about one-third patients with epilepsy exhibits resistance to available antiseizures medications, known as drug-resistant epilepsy (DRE). Mitochondrial dysfunction has been implicated as a hallmark in drug-resistant epilepsy via activation of microglial kynurenine 3-monooxygenase (KMO) and cyclooxygenase (COX) enzymes, leading to neuroinflammation and oxidative stress. Diclofenac, an equipotent non selective cyclooxygenase inhibitor, has inhibitory action on KMO enzyme and has also shown anti-inflammatory and antioxidant properties in animal models of epilepsy. These properties make it a suitable candidate for amelioration of DRE. However, its potential in drug-resistant epilepsy remained unexplored till date. In this study, dose dependent effect of diclofenac (5 mg/kg, 10 mg/kg, 20 mg/kg) has been explored in rotenone corneal kindling model of mitochondrial DRE. The results of our study revealed the induction of drug resistance to antiseizure medications and induced kynurenine 3-monooxygenase activity in rotenone corneal kindled epileptic mice in comparison to naive mice. Treatment of rotenone corneal kindled epileptic mice with diclofenac resulted in a significant decrease in drug resistance to antiseizure medications as evident by a reduction in seizure score in the treatment groups as compared to control group, in post-treatment resistance validation. The kynurenine 3-monooxygenase inhibitory activity (as evidenced by decreased levels of neurotoxic quinolinic acid) and the antioxidant effect (as evident by significantly reduced oxidative stress) in the diclofenac treated groups, emerged as a major contributor for its ameliorative action. Findings of this study suggests, diclofenac can be used as an adjunct therapy in amelioration of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Samriti
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - Arvinder Kaur
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - Arshbir Kaur
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - R K Goel
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| |
Collapse
|
3
|
Yang JY, Shin DS, Jeong M, Kim SS, Jeong HN, Lee BH, Hwang KS, Son Y, Jeong HC, Choi CH, Lee KR, Bae MA. Evaluation of Drug Blood-Brain-Barrier Permeability Using a Microfluidic Chip. Pharmaceutics 2024; 16:574. [PMID: 38794236 PMCID: PMC11125197 DOI: 10.3390/pharmaceutics16050574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 05/26/2024] Open
Abstract
The blood-brain-barrier (BBB) is made up of blood vessels whose permeability enables the passage of some compounds. A predictive model of BBB permeability is important in the early stages of drug development. The predicted BBB permeabilities of drugs have been confirmed using a variety of in vitro methods to reduce the quantities of drug candidates needed in preclinical and clinical trials. Most prior studies have relied on animal or cell-culture models, which do not fully recapitulate the human BBB. The development of microfluidic models of human-derived BBB cells could address this issue. We analyzed a model for predicting BBB permeability using the Emulate BBB-on-a-chip machine. Ten compounds were evaluated, and their permeabilities were estimated. Our study demonstrated that the permeability trends of ten compounds in our microfluidic-based system resembled those observed in previous animal and cell-based experiments. Furthermore, we established a general correlation between the partition coefficient (Kp) and the apparent permeability (Papp). In conclusion, we introduced a new paradigm for predicting BBB permeability using microfluidic-based systems.
Collapse
Affiliation(s)
- Jung Yoon Yang
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Dae-Seop Shin
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Moonkyu Jeong
- Department of Bioengineering, University of Science and Technology, Daejeon 34113, Republic of Korea;
- Laboratory Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Seong Soon Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Ha Neul Jeong
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Byung Hoi Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Kyu-Seok Hwang
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Yuji Son
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
| | - Hyeon-Cheol Jeong
- Laboratory Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Chi-Hoon Choi
- Department of Radiology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea;
| | - Kyeong-Ryoon Lee
- Department of Bioengineering, University of Science and Technology, Daejeon 34113, Republic of Korea;
- Laboratory Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Myung Ae Bae
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; (J.Y.Y.); (D.-S.S.); (S.S.K.); (H.N.J.); (B.H.L.); (K.-S.H.); (Y.S.)
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
4
|
Vázquez M, Fagiolino P. The role of efflux transporters and metabolizing enzymes in brain and peripheral organs to explain drug-resistant epilepsy. Epilepsia Open 2021; 7 Suppl 1:S47-S58. [PMID: 34560816 PMCID: PMC9340310 DOI: 10.1002/epi4.12542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 11/08/2022] Open
Abstract
Drug‐resistant epilepsy has been explained by different mechanisms. The most accepted one involves overexpression of multidrug transporters proteins at the blood brain barrier and brain metabolizing enzymes. This hypothesis is one of the main pharmacokinetic reasons that lead to the lack of response of some antiseizure drug substrates of these transporters and enzymes due to their limited entrance into the brain and limited stay at the sites of actions. Although uncontrolled seizures can be the cause of the overexpression, some antiseizure medications themselves can cause such overexpression leading to treatment failure and thus refractoriness. However, it has to be taken into account that the inductive effect of some drugs such as carbamazepine or phenytoin not only impacts on the brain but also on the rest of the body with different intensity, influencing the amount of drug available for the central nervous system. Such induction is not only local drug concentration but also time dependent. In the case of valproic acid, the deficient disposition of ammonia due to a malfunction of the urea cycle, which would have its origin in an intrinsic deficiency of L‐carnitine levels in the patient or by its depletion caused by the action of this antiseizure drug, could lead to drug‐resistant epilepsy. Many efforts have been made to change this situation. In order to name some, the administration of once‐daily dosing of phenytoin or the coadministration of carnitine with valproic acid would be preferable to avoid iatrogenic refractoriness. Another could be the use of an adjuvant drug that down‐regulates the expression of transporters. In this case, the use of cannabidiol with antiseizure properties itself and able to diminish the overexpression of these transporters in the brain could be a novel therapy in order to allow penetration of other antiseizure medications into the brain.
Collapse
Affiliation(s)
- Marta Vázquez
- Pharmaceutical Sciences Department, Faculty of Chemistry, Universidad de la República, Montevideo, Uruguay
| | - Pietro Fagiolino
- Pharmaceutical Sciences Department, Faculty of Chemistry, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
5
|
Sheng Y, Yang H, Wu T, Zhu L, Liu L, Liu X. Alterations of Cytochrome P450s and UDP-Glucuronosyltransferases in Brain Under Diseases and Their Clinical Significances. Front Pharmacol 2021; 12:650027. [PMID: 33967789 PMCID: PMC8097730 DOI: 10.3389/fphar.2021.650027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Cytochrome P450s (CYPs) and UDP-glucuronosyltransferases (UGTs) are both greatly important metabolic enzymes in various tissues, including brain. Although expressions of brain CYPs and UGTs and their contributions to drug disposition are much less than liver, both CYPs and UGTs also mediate metabolism of endogenous substances including dopamine and serotonin as well as some drugs such as morphine in brain, demonstrating their important roles in maintenance of brain homeostasis or pharmacological activity of drugs. Some diseases such as epilepsy, Parkinson's disease and Alzheimer's disease are often associated with the alterations of CYPs and UGTs in brain, which may be involved in processes of these diseases via disturbing metabolism of endogenous substances or resisting drugs. This article reviewed the alterations of CYPs and UGTs in brain, the effects on endogenous substances and drugs and their clinical significances. Understanding the roles of CYPs and UGTs in brain provides some new strategies for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Yun Sheng
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Yang
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tong Wu
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Zhu
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li Liu
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Pharmacokinetics and Metabolism, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Williams-Medina A, Deblock M, Janigro D. In vitro Models of the Blood-Brain Barrier: Tools in Translational Medicine. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 2:623950. [PMID: 35047899 PMCID: PMC8757867 DOI: 10.3389/fmedt.2020.623950] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/28/2022] Open
Abstract
Medical progress has historically depended on scientific discoveries. Until recently, science was driven by technological advancements that, once translated to the clinic, fostered new treatments and interventions. More recently, technology-driven medical progress has often outpaced laboratory research. For example, intravascular devices, pacemakers for the heart and brain, spinal cord stimulators, and surgical robots are used routinely to treat a variety of diseases. The rapid expansion of science into ever more advanced molecular and genetic mechanisms of disease has often distanced laboratory-based research from day-to-day clinical realities that remain based on evidence and outcomes. A recognized reason for this hiatus is the lack of laboratory tools that recapitulate the clinical reality faced by physicians and surgeons. To overcome this, the NIH and FDA have in the recent past joined forces to support the development of a "human-on-a-chip" that will allow research scientists to perform experiments on a realistic replica when testing the effectiveness of novel experimental therapies. The development of a "human-on-a-chip" rests on the capacity to grow in vitro various organs-on-a-chip, connected with appropriate vascular supplies and nerves, and our ability to measure and perform experiments on these virtually invisible organs. One of the tissue structures to be scaled down on a chip is the human blood-brain barrier. This review gives a historical perspective on in vitro models of the BBB and summarizes the most recent 3D models that attempt to fill the gap between research modeling and patient care. We also present a summary of how these in vitro models of the BBB can be applied to study human brain diseases and their treatments. We have chosen NeuroAIDS, COVID-19, multiple sclerosis, and Alzheimer's disease as examples of in vitro model application to neurological disorders. Major insight pertaining to these illnesses as a consequence of more profound understanding of the BBB can reveal new avenues for the development of diagnostics, more efficient therapies, and definitive clarity of disease etiology and pathological progression.
Collapse
Affiliation(s)
- Alberto Williams-Medina
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States
- Flocel, Inc., Cleveland, OH, United States
| | - Michael Deblock
- Department of Biomedical Engineering, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Damir Janigro
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States
- Flocel, Inc., Cleveland, OH, United States
| |
Collapse
|
7
|
Heat Shock Proteins Accelerate the Maturation of Brain Endothelial Cell Glucocorticoid Receptor in Focal Human Drug-Resistant Epilepsy. Mol Neurobiol 2020; 57:4511-4529. [PMID: 32748370 DOI: 10.1007/s12035-020-02043-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/24/2020] [Indexed: 12/21/2022]
Abstract
Pharmacoresistance in epilepsy is a major challenge to successful clinical therapy. Glucocorticoid receptor (GR) dysregulation can affect the underlying disease pathogenesis. We recently reported that local drug biotransformation at the blood-brain barrier is upregulated by GR, which controls drug-metabolizing enzymes (e.g., cytochrome P450s, CYPs) and efflux drug transporters (MDR1) in human epileptic brain endothelial cells (EPI-ECs). Here, we establish that this mechanism is influenced upstream by GR and its association with heat shock proteins/co-chaperones (Hsps) during maturation, which differentially affect human epileptic (EPI) tissue and brain endothelial cells. Overexpressed GR, Hsp90, Hsp70, and Hsp40 were found in EPI vs. NON-EPI brain regions. Elevated neurovascular GR expression and co-localization with Hsps was evident in the EPI regions with cortical dysplasia, predominantly in the brain micro-capillaries and neurons. A corresponding increase in ATPase activity (*p < 0.05) was found in the EPI regions. The GR-Hsp90/Hsp70 binding patterns indicated a faster chaperone-promoted maturation of GR, leading to its overactivation in both the tissue and EPI-ECs derived from EPI/focal regions and GR silencing in EPI-ECs slowed such GR-Hsp interactions. Significantly accelerated GR nuclear translocation was determined in EPI-ECs following treatment with GR modulators/ligands dexamethasone, rifampicin, or phenytoin. Our findings reveal that overexpressed GR co-localizes with Hsps in the neurovasculature of EPI brain, increased GR maturation by Hsps accelerates EPI GR machinery, and furthermore this change in EPI and NON-EPI GR-Hsp interaction alters with the age of seizure onset in epileptic patients, together affecting the pathophysiology and drug regulation in the epileptic brain endothelium.
Collapse
|
8
|
Williams S, Hossain M, Ferguson L, Busch RM, Marchi N, Gonzalez-Martinez J, Perucca E, Najm IM, Ghosh C. Neurovascular Drug Biotransformation Machinery in Focal Human Epilepsies: Brain CYP3A4 Correlates with Seizure Frequency and Antiepileptic Drug Therapy. Mol Neurobiol 2019; 56:8392-8407. [PMID: 31243719 DOI: 10.1007/s12035-019-01673-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/07/2019] [Indexed: 01/01/2023]
Abstract
Pharmacoresistance is a major clinical challenge for approximately 30% of patients with epilepsy. Previous studies indicate nuclear receptors (NRs), drug efflux transporters, and cytochrome P450 enzymes (CYPs) control drug passage across the blood-brain barrier (BBB) in drug-resistant epilepsy. Here, we (1) evaluate BBB changes, neurovascular nuclear receptors, and drug transporters in lesional/epileptic (EPI) and non-lesional/non-epileptic (NON-EPI) regions of the same brain, (2) examine regional CYP expression and activity, and (3) investigate the association among CYP brain expression, seizure frequency, duration of epilepsy, and antiepileptic drug (AED) combination. We used surgically resected brain specimens from patients with medically intractable epilepsy (n = 22) where the epileptogenic loci were well-characterized by invasive and non-invasive methods; histology confirmed distinction of small NON-EPI regions from EPI tissues. NRs, transporters, CYPs, and tight-junction proteins were assessed by western blots/immunohistochemistry, and CYP metabolic activity was determined and compared. The relationship of CYP expression with seizure frequency, duration of epilepsy, and prescribed AEDs was evaluated. Decreased BBB tight-junction proteins accompanied IgG leakage in EPI regions and correlated with upregulated NR and efflux transporter levels. CYP expression and activity significantly increased in EPI compared to NON-EPI tissues. Change in EPI and NON-EPI CYP3A4 expression increased in patients taking AEDs that were CYP substrates, was downregulated when CYP- and non-CYP-substrate AEDs were given together, and correlated with seizure frequency. Our studies suggest focal neurovascular CYP-NR-transporter alterations, as demonstrated by the relationship of seizure frequency and AED combination to brain CYP3A4, might together impact biotransformation machinery of human pharmacoresistant epilepsy.
Collapse
Affiliation(s)
- Sherice Williams
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Mohammed Hossain
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Lisa Ferguson
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robyn M Busch
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain Disorders Laboratory, Department of Neuroscience, Institute of Functional Genomics (CNRS-INSERM), University of Montpellier, Montpellier, France
| | | | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia, Clinical Trial Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Imad M Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chaitali Ghosh
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
9
|
Ghosh C, Hossain M, Mishra S, Khan S, Gonzalez-Martinez J, Marchi N, Janigro D, Bingaman W, Najm I. Modulation of glucocorticoid receptor in human epileptic endothelial cells impacts drug biotransformation in an in vitro blood-brain barrier model. Epilepsia 2018; 59:2049-2060. [PMID: 30264400 PMCID: PMC6282717 DOI: 10.1111/epi.14567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 12/24/2022]
Abstract
Objective Nuclear receptors and cytochrome P450 (CYP) regulate hepatic metabolism of several drugs. Nuclear receptors are expressed at the neurovascular unit of patients with drug‐resistant epilepsy. We studied whether glucocorticoid receptor (GR) silencing or inhibition in human epileptic brain endothelial cells (EPI‐ECs) functionally impacts drug bioavailability across an in vitro model of the blood–brain barrier (BBB) by CYP‐multidrug transporter (multidrug resistance protein 1, MDR1) mechanisms. Methods Surgically resected brain specimens from patients with drug‐resistant epilepsy, primary EPI‐ECs, and control human brain microvascular endothelial cells (HBMECs) were used. Expression of GR, pregnane X receptor, CYP3A4, and MDR1 was analyzed pre‐ and post‐GR silencing in EPI‐ECs. Endothelial cells were co‐cultured with astrocytes and seeded in an in vitro flow‐based BBB model (DIV‐BBB). Alternatively, the GR inhibitor mifepristone was added to the EPI‐EC DIV‐BBB. Integrity of the BBB was monitored by measuring transendothelial electrical resistance. Cell viability was assessed by glucose‐lactate levels. Permeability of [3H]sucrose and [14C]phenytoin was quantified. CYP function was determined by measuring resorufin formation and oxcarbazepine (OXC) metabolism. Results Silencing and inhibition of GR in EPI‐ECs resulted in decreased pregnane X receptor, CYP3A4, and MDR1 expression. GR silencing or inhibition did not affect BBB properties in vitro, as transendothelial electrical resistance and Psucrose were unaltered, and glucose metabolism was maintained. GR EPI‐EC silencing or inhibition led to (1) increased PphenytoinBBB permeability as compared to control; (2) decreased CYP function, indirectly evaluated by resorufin formation; (3) improved OXC bioavailability with increased abluminal (brain‐side) OXC levels as compared to control. Significance Our results suggest that modulating GR expression in EPI‐ECs at the BBB modifies drug metabolism and penetration by a mechanism encompassing P450 and efflux transporters. The latter could be exploited for future drug design and to overcome pharmacoresistance.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, Ohio
| | - Mohammed Hossain
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Saurabh Mishra
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sameena Khan
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain Disorders Laboratory, Department of Neuroscience, Institute of Functional Genomics (CNRS-INSERM), University of Montpellier, Montpellier, France
| | - Damir Janigro
- Flocel, Inc., Cleveland, Ohio.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| | - William Bingaman
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Imad Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
10
|
Ghosh C, Hossain M, Solanki J, Najm IM, Marchi N, Janigro D. Overexpression of pregnane X and glucocorticoid receptors and the regulation of cytochrome P450 in human epileptic brain endothelial cells. Epilepsia 2017; 58:576-585. [PMID: 28199000 PMCID: PMC5386820 DOI: 10.1111/epi.13703] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2017] [Indexed: 01/31/2023]
Abstract
Objective Recent evidence suggests a metabolic contribution of cytochrome P450 enzymes (CYPs) to the drug‐resistant phenotype in human epilepsy. However, the upstream molecular regulators of CYP in the epileptic brain remain understudied. We therefore investigated the expression and function of pregnane xenobiotic (PXR) and glucocorticoid (GR) nuclear receptors in endothelial cells established from post‐epilepsy surgery brain samples. Methods PXR/GR localization was evaluated by immunohistochemistry in specimens from subjects who underwent temporal lobe resections to relieve drug‐resistant seizures. We used primary cultures of endothelial cells obtained from epileptic brain tissues (EPI‐ECs; n = 8), commercially available human brain microvascular endothelial cells (HBMECs; n = 8), and human hepatocytes (n = 3). PXR/GR messenger RNA (mRNA) levels in brain ECs was initially determined by complementary DNA (cDNA) microarrays. The expression of PXR/GR proteins was quantified by Western blot. PXR and GR silencing was performed in EPI‐ECs (n = 4), and the impact on downstream CYP expression was determined. Results PXR/GR expression was detected by immunofluorescence in ECs and neurons in the human temporal lobe samples analyzed. Elevated mRNA and protein levels of PXR and GR were found in EPI‐ECs versus control HBMECs. Hepatocytes, used as a positive control, displayed the highest levels of PXR/GR expression. We confirmed expression of PXR/GR in cytoplasmic‐nuclear subcellular fractions, with a significant increase of PXR/GR in EPI‐ECs versus controls. CYP3A4, CYP2C9, and CYP2E1 were overexpressed in EPI‐ECs versus control, whereas CYP2D6 and CYP2C19 were downregulated or absent in EPI‐ECs. GR silencing in EPI‐ECs led to decreased CYP3A4, CYP2C9, and PXR expression. PXR silencing in EPI‐ECs resulted in the specific downregulation of CYP3A4 expression. Significance Our results indicate increased PXR and GR in primary ECs derived from human epileptic brains. PXR or GR may be responsible for a local drug brain metabolism sustained by abnormal CYP regulation.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A.,Department of Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Mohammed Hossain
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Jesal Solanki
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Imad M Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, U.S.A
| | - Nicola Marchi
- Department of Neuroscience, Institute of Functional Genomics, CNRS/INSERM, Montpellier, France
| | - Damir Janigro
- Flocel, Inc., Cleveland, Ohio, U.S.A.,Case Western Reserve University, Cleveland, Ohio, U.S.A
| |
Collapse
|
11
|
Ghosh C, Hossain M, Solanki J, Dadas A, Marchi N, Janigro D. Pathophysiological implications of neurovascular P450 in brain disorders. Drug Discov Today 2016; 21:1609-1619. [PMID: 27312874 DOI: 10.1016/j.drudis.2016.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/14/2016] [Accepted: 06/06/2016] [Indexed: 01/12/2023]
Abstract
Over the past decades, the significance of cytochrome P450 (CYP) enzymes has expanded beyond their role as peripheral drug metabolizers in the liver and gut. CYP enzymes are also functionally active at the neurovascular interface. CYP expression is modulated by disease states, impacting cellular functions, detoxification, and reactivity to toxic stimuli and brain drug biotransformation. Unveiling the physiological and molecular complexity of brain P450 enzymes will improve our understanding of the mechanisms underlying brain drug availability, pharmacological efficacy, and neurotoxic adverse effects from pharmacotherapy targeting brain disorders.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| | - Mohammed Hossain
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | | | - Aaron Dadas
- The Ohio State University, Columbus, OH, USA
| | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (CNRS/INSERM), Montpellier, France
| | - Damir Janigro
- Flocel Inc. and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
12
|
Daci A, Beretta G, Vllasaliu D, Shala A, Govori V, Norata GD, Krasniqi S. Polymorphic Variants of SCN1A and EPHX1 Influence Plasma Carbamazepine Concentration, Metabolism and Pharmacoresistance in a Population of Kosovar Albanian Epileptic Patients. PLoS One 2015; 10:e0142408. [PMID: 26555147 PMCID: PMC4640545 DOI: 10.1371/journal.pone.0142408] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/20/2015] [Indexed: 12/24/2022] Open
Abstract
Aim The present study aimed to evaluate the effects of gene variants in key genes influencing pharmacokinetic and pharmacodynamic of carbamazepine (CBZ) on the response in patients with epilepsy. Materials & Methods Five SNPs in two candidate genes influencing CBZ transport and metabolism, namely ABCB1 or EPHX1, and CBZ response SCN1A (sodium channel) were genotyped in 145 epileptic patients treated with CBZ as monotherapy and 100 age and sex matched healthy controls. Plasma concentrations of CBZ, carbamazepine-10,11-epoxide (CBZE) and carbamazepine-10,11-trans dihydrodiol (CBZD) were determined by HPLC-UV-DAD and adjusted for CBZ dosage/kg of body weight. Results The presence of the SCN1A IVS5-91G>A variant allele is associated with increased epilepsy susceptibility. Furthermore, carriers of the SCN1A IVS5-91G>A variant or of EPHX1 c.337T>C variant presented significantly lower levels of plasma CBZ compared to carriers of the common alleles (0.71±0.28 vs 1.11±0.69 μg/mL per mg/Kg for SCN1A IVS5-91 AA vs GG and 0.76±0.16 vs 0.94±0.49 μg/mL per mg/Kg for EPHX1 c.337 CC vs TT; P<0.05 for both). Carriers of the EPHX1 c.416A>G showed a reduced microsomal epoxide hydrolase activity as reflected by a significantly decreased ratio of CBZD to CBZ (0.13±0.08 to 0.26±0.17, p<0.05) also of CBZD to CBZE (1.74±1.06 to 3.08±2.90; P<0.05) and CDRCBZD (0.13±0.08 vs 0.24±0.19 μg/mL per mg/Kg; P<0.05). ABCB1 3455C>T SNP and SCN1A 3148A>G variants were not associated with significant changes in CBZ pharmacokinetic. Patients resistant to CBZ treatment showed increased dosage of CBZ (657±285 vs 489±231 mg/day; P<0.001) but also increased plasma levels of CBZ (9.84±4.37 vs 7.41±3.43 μg/mL; P<0.001) compared to patients responsive to CBZ treatment. CBZ resistance was not related to any of the SNPs investigated. Conclusions The SCN1A IVS5-91G>A SNP is associated with susceptibility to epilepsy. SNPs in EPHX1 gene are influencing CBZ metabolism and disposition. CBZ plasma levels are not an indicator of resistance to the therapy.
Collapse
Affiliation(s)
- Armond Daci
- Department of Pharmacy, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
- Institute of Pharmacology and Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giangiacomo Beretta
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Driton Vllasaliu
- University of Lincoln, School of Pharmacy, Joseph Banks Laboratories, Green Lane, Lincoln, LN6 7DL, United Kingdom
| | - Aida Shala
- Department of Pharmacy, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Valbona Govori
- Neurology Clinic, University Clinical Center of Kosova, Prishtina, Kosovo
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Shaip Krasniqi
- Institute of Pharmacology and Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
- * E-mail:
| |
Collapse
|
13
|
Ghosh C, Hossain M, Spriggs A, Ghosh A, Grant GA, Marchi N, Perucca E, Janigro D. Sertraline-induced potentiation of the CYP3A4-dependent neurotoxicity of carbamazepine: an in vitro study. Epilepsia 2015; 56:439-49. [PMID: 25656284 PMCID: PMC4413932 DOI: 10.1111/epi.12923] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Drug toxicity is a hurdle to drug development and to clinical translation of basic research. Antiepileptic drugs such as carbamazepine (CBZ) and selective serotonin reuptake inhibitors such as sertraline (SRT) are commonly co-prescribed to patients with epilepsy and comorbid depression. Because SRT may interfere with cytochrome P450 (CYP) enzyme activity and CYPs have been implicated in the conversion of CBZ to reactive cytotoxic metabolites, we investigated in vitro models to determine whether SRT affects the neurotoxic potential of CBZ and the mechanisms involved. METHODS Human fetal brain-derived dopaminergic neurons, human brain microvascular endothelial cells (HBMECs), and embryonic kidney (HEK) cells were used to evaluate cytotoxicity of CBZ and SRT individually and in combination. Nitrite and glutathione (GSH) levels were measured with drug exposure. To validate the role of CYP3A4 in causing neurotoxicity, drug metabolism was compared to cell death in HEK CYP3A4 overexpressed and cells pretreated with the CYP3A4 inhibitor ketoconazole. RESULTS In all cellular systems tested, exposure to CBZ (127 μM) or SRT (5 μM) alone caused negligible cytotoxicity. By contrast CBZ, tested at a much lower concentration (17 μM) in combination with SRT (5 μM), produced prominent cytotoxicity within 15 min exposure. In neurons and HBMECs, cytotoxicity was associated with increased nitrite levels, suggesting involvement of free radicals as a pathogenetic mechanism. Pretreatment of HBMECs with reduced GSH or with the GSH precursor N-acetyl-L-cysteine prevented cytotoxic response. In HEK cells, the cytotoxic response to the CBZ + SRT combination correlated with the rate of CBZ biotransformation and production of 2-hydroxy CBZ, further suggesting a causative role of reactive metabolites. In the same system, cytotoxicity was potentiated by overexpression of CYP3A4, and prevented by CYP3A4 inhibitor. SIGNIFICANCE These results demonstrate an unexpected neurotoxic interaction between CBZ and SRT, apparently related to increased CYP3A4-mediated production of reactive CBZ metabolites. The potential clinical implications of these findings are discussed.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Bio-medical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
| | - Mohammad Hossain
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Bio-medical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
| | - Addison Spriggs
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Bio-medical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
| | - Arnab Ghosh
- Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
| | - Gerald A. Grant
- Department of Neurosurgery and Neurobiology, Stanford University School of Medicine, Stanford, California, U.S.A
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics, CNRS, Montpellier, France
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia and C. Mondino National Neurological Institute, Pavia, Italy
| | - Damir Janigro
- Cerebrovascular Research, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Bio-medical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
- Department of Neurosurgery, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, U.S.A
| |
Collapse
|
14
|
Sandow N, Kim S, Raue C, Päsler D, Klaft ZJ, Antonio LL, Hollnagel JO, Kovacs R, Kann O, Horn P, Vajkoczy P, Holtkamp M, Meencke HJ, Cavalheiro EA, Pragst F, Gabriel S, Lehmann TN, Heinemann U. Drug resistance in cortical and hippocampal slices from resected tissue of epilepsy patients: no significant impact of p-glycoprotein and multidrug resistance-associated proteins. Front Neurol 2015; 6:30. [PMID: 25741317 PMCID: PMC4332373 DOI: 10.3389/fneur.2015.00030] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/03/2015] [Indexed: 01/16/2023] Open
Abstract
Drug resistant patients undergoing epilepsy surgery have a good chance to become sensitive to anticonvulsant medication, suggesting that the resected brain tissue is responsible for drug resistance. Here, we address the question whether P-glycoprotein (Pgp) and multidrug resistance-associated proteins (MRPs) expressed in the resected tissue contribute to drug resistance in vitro. Effects of anti-epileptic drugs [carbamazepine (CBZ), sodium valproate, phenytoin] and two unspecific inhibitors of Pgp and MRPs [verapamil (VPM) and probenecid (PBN)] on seizure-like events (SLEs) induced in slices from 35 hippocampal and 35 temporal cortex specimens of altogether 51 patients (161 slices) were studied. Although in slice preparations the blood brain barrier is not functional, we found that SLEs predominantly persisted in the presence of anticonvulsant drugs (90%) and also in the presence of VPM and PBN (86%). Following subsequent co-administration of anti-epileptic drugs and drug transport inhibitors, SLEs continued in 63% of 143 slices. Drug sensitivity in slices was recognized either as transition to recurrent epileptiform transients (30%) or as suppression (7%), particularly by perfusion with CBZ in PBN containing solutions (43, 9%). Summarizing responses to co-administration from more than one slice per patient revealed that suppression of seizure-like activity in all slices was only observed in 7% of patients. Patients whose tissue was completely or partially sensitive (65%) presented with higher seizure frequencies than those with resistant tissue (35%). However, corresponding subgroups of patients do not differ with respect to expression rates of drug transporters. Our results imply that parenchymal MRPs and Pgp are not responsible for drug resistance in resected tissue.
Collapse
Affiliation(s)
- Nora Sandow
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany ; Department of Neurosurgery, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Simon Kim
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Claudia Raue
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Dennis Päsler
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Zin-Juan Klaft
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Leandro Leite Antonio
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany ; Laboratorio de Neurologia Experimental, Universidade Federal de São Paulo-Escola Paulista de Medicina , São Paulo , Brazil
| | - Jan Oliver Hollnagel
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Richard Kovacs
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Oliver Kann
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany ; Institute of Physiology and Pathophysiology, University of Heidelberg , Heidelberg , Germany
| | - Peter Horn
- Department of Neurosurgery, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Martin Holtkamp
- Epilepsy Center of Berlin-Brandenburg, Ev. Krankenhaus Königin Elisabeth Herzberge , Berlin , Germany
| | - Heinz-Joachim Meencke
- Epilepsy Center of Berlin-Brandenburg, Ev. Krankenhaus Königin Elisabeth Herzberge , Berlin , Germany
| | - Esper A Cavalheiro
- Laboratorio de Neurologia Experimental, Universidade Federal de São Paulo-Escola Paulista de Medicina , São Paulo , Brazil
| | - Fritz Pragst
- Institute of Forensic Medicine - Forensic Toxicology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | - Siegrun Gabriel
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| | | | - Uwe Heinemann
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
15
|
Effect of status epilepticus and antiepileptic drugs on CYP2E1 brain expression. Neuroscience 2014; 281:124-34. [PMID: 25280786 DOI: 10.1016/j.neuroscience.2014.09.055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/01/2014] [Accepted: 09/25/2014] [Indexed: 10/24/2022]
Abstract
P450 metabolic enzymes are expressed in the human and rodent brain. Recent data support their involvement in the pathophysiology of epilepsy. However, the determinants of metabolic enzyme expression in the epileptic brain are unclear. We tested the hypothesis that status epilepticus (SE) or exposure to phenytoin or phenobarbital affects brain expression of the metabolic enzyme CYP2E1. SE was induced in C57BL/6J mice by systemic kainic acid. Brain CYP2E1 expression was evaluated 18-24h after severe SE by immunohistochemistry. Co-localization with neuronal nuclei (NEUN), glial fibrillary acidic protein (GFAP) and CD31 was determined by confocal microscopy. The effect of phenytoin, carbamazepine and phenobarbital on CYP2E1 expression was evaluated in vivo or by using organotypic hippocampal cultures in vitro. CYP2E1 expression was investigated in brain resections from a cohort of drug-resistant epileptic brain resections and human endothelial cultures (EPI-EC). Immunohistochemistry showed an increase of CYP2E1 expression limited to hippocampal CA2/3 and hilar neurons after severe SE in mice. CYP2E1 expression was also observed at the astrocyte-vascular interface. Analysis of human brain specimens revealed CYP2E1 expression in neurons and vascular endothelial cells (EC). CYP2E1 was expressed in cultured human EC and over-expressed by EPI-EC. When analyzing the effect of drug exposure on CYP2E1 expression we found that, in vivo or in vitro, ethanol increased CYP2E1 levels in the brain and liver. Treatment with phenytoin induced localized CYP2E1 expression in the brain whereas no significant effects were exerted by carbamazepine or phenobarbital. Our data indicate that the effect of acute SE on brain CYP2E1 expression is localized and cell specific. Exposure to selected anti-epileptic drugs could play a role in determining CYP2E1 brain expression. Additional investigation is required to fully reproduce the culprits of P450 enzyme expression as observed in the human epileptic brain.
Collapse
|
16
|
Marasco CC, Goodwin CR, Winder DG, Schramm-Sapyta NL, McLean JA, Wikswo JP. Systems-level view of cocaine addiction: the interconnection of the immune and nervous systems. Exp Biol Med (Maywood) 2014; 239:1433-42. [PMID: 24903164 DOI: 10.1177/1535370214537747] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The human body is a complex assembly of physiological systems designed to manage the multidirectional transport of both information and nutrients. An intricate interplay between the nervous, circulatory, and secretory systems is therefore necessary to sustain life, allow delivery of nutrients and therapeutic drugs, and eliminate metabolic waste products and toxins. These systems also provide vulnerable routes for modification by substances of abuse. Addictive substances are, by definition, neurologically active, but as they and their metabolites are spread throughout the body via the nervous, circulatory, respiratory and digestive systems, there is abundant opportunity for interaction with numerous cell and tissue types. Cocaine is one such substance that exerts a broad physiological effect. While a great deal of the research concerning addiction has addressed the neurological effects of cocaine use, only a few studies have been aimed at delineating the role that cocaine plays in various body systems. In this paper, we probe the current research regarding cocaine and the immune system, and map a systems-level view to outline a broader perspective of the biological response to cocaine. Specifically, our overview of the neurological and immunomodulatory effects of the drug will allow a broader perspective of the biological response to cocaine. The focus of this review is on the connection between the nervous and immune systems and the role this connection plays in the long-term complications of cocaine use. By describing the multiplicity of these connections, we hope to inspire detailed investigations into the immunological interplay in cocaine addiction.
Collapse
Affiliation(s)
- Christina C Marasco
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Cody R Goodwin
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | | | - John A McLean
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - John P Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
17
|
Ghosh C, Hossain M, Puvenna V, Martinez-Gonzalez J, Alexopolous A, Janigro D, Marchi N. Expression and functional relevance of UGT1A4 in a cohort of human drug-resistant epileptic brains. Epilepsia 2013; 54:1562-70. [PMID: 23865846 DOI: 10.1111/epi.12318] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Brain drug bioavailability is regulated by the blood-brain barrier (BBB). It was recently suggested that cytochrome P450 (CYP) enzymes could act in concert with multidrug transporter proteins to regulate drug penetration and distribution into the diseased brain. The possibility that phase II metabolic enzymes could be expressed in the epileptic brain has been not evaluated. Phase II enzymes are involved in the metabolism of common antiepileptic drugs (AEDs). METHODS Phase II enzyme UGT1A4 brain expression was evaluated in temporal lobe resections from patients with epilepsy. UGT1A4 expression was determined by western blot and immunocytochemistry in primary cultures of human drug-resistant brain endothelial human brain epileptic endothelial cells (EPI-EC)s and commercially available control cells human brain microvascular endothelial cells (HBMECs). Lack of DNA condensation measured by 4',6-diamidino-2-phenylindole (DAPI) was used as a surrogate marker of cell viability and was correlated to UGT1A4 expression high performance liquid chromatography ultraviolet detection (HPLC-UV) was used to quantify lamotrigine metabolism by EPI-EC and HBMEC. The appearance of the specific lamotrigine metabolite, 2-n glucuronide (MET-1), was also evaluated. Lamotrigine and MET-1 levels were measured in selected surgical brain and matched blood samples. KEY FINDINGS UGT1A4 expression was observed in BBB endothelial cells and neurons. Our quantification study revealed variable levels of UGT1A4 expression across the brain specimens analyzed. Neurons devoid of UGT1A4 expression displayed nuclear DAPI condensation, a sign of cellular distress. UGT1A4 overexpression in EPI-EC, as compared to HBMEC, was reflected by a proportional increase in lamotrigine metabolism. The lamotrigine metabolite, MET-1, was formed in vitro by EPI-EC and, to a lesser extent, by HBMEC. HPLC-UV measurements of brain and blood samples obtained from patients receiving lamotrigine prior to surgery revealed the presence of lamotrigine and its metabolites in the brain. SIGNIFICANCE These initial results suggest the presence of a phase II enzyme in the epileptic brain. Further studies are required to fully describe the pattern of brain UGT1A4 expression in relation to clinical variables and drug resistance.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Department of Cellular, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Cucullo L, Hossain M, Tierney W, Janigro D. A new dynamic in vitro modular capillaries-venules modular system: cerebrovascular physiology in a box. BMC Neurosci 2013; 14:18. [PMID: 23388041 PMCID: PMC3598202 DOI: 10.1186/1471-2202-14-18] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 01/25/2013] [Indexed: 01/30/2023] Open
Abstract
Background The study of the cerebrovascular physiology is crucial to understand the pathogenesis of neurological disease and the pharmacokinetic of drugs. Appropriate models in vitro often fail to represent in vivo physiology. To address these issues we propose the use of a novel artificial vascular system that closely mimics capillary and venous segments of human cerebrovasculature while also allowing for an extensive control of the experimental variables and their manipulation. Results Using hollow fiber technology, we modified an existing dynamic artificial model of the blood–brain barrier (BBB) (DIV-capillary) to encompass the distal post-capillary (DIV-venules) segments of the brain circulatory system. This artificial brain vascular system is comprised of a BBB module serially connected to a venule segment. A pump generates a pulsatile flow with arterial pressure feeding the system. The perfusate of the capillary module achieves levels of shear stress, pressure, and flow rate comparable to what observed in situ. Endothelial cell exposure to flow and abluminal astrocytic stimuli allowed for the formation of a highly selective capillary BBB with a trans-endothelial electrical resistance (TEER; >700 ohm cm2) and sucrose permeability (< 1X10-u cm/sec) comparable to in vivo. The venule module, which attempted to reproduce features of the hemodynamic microenvironment of venules, was perfused by media resulting in shear stress and intraluminal pressure levels lower than those found in capillaries. Because of altered cellular and hemodynamic factors, venule segments present a less stringent vascular bed (TEER <250 Ohm cm2; Psucrose > 1X10-4 cm/sec) than that of the BBB. Abluminal human brain vascular smooth muscle cells were used to reproduce the venular abluminal cell composition. Conclusion The unique characteristics afforded by the DIV-BBB in combination with a venule segment will realistically expand our ability to dissect and study the physiological and functional behavior of distinct segments of the human cerebrovascular network.
Collapse
Affiliation(s)
- Luca Cucullo
- Cerebrovascular Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|