1
|
Eddy K, Gupta K, Pelletier JC, Isola AL, Marinaro C, Rasheed MA, Campagnolo J, Eddin MN, Rossi M, Fateeva A, Reuhl K, Shah R, Robinson AK, Chaly A, Freeman KB, Chen W, Diaz J, Furmanski P, Silk AW, Reitz AB, Zloza A, Chen S. A Spontaneous Melanoma Mouse Model Applicable for a Longitudinal Chemotherapy and Immunotherapy Study. J Invest Dermatol 2023; 143:2007-2018.e6. [PMID: 36997110 PMCID: PMC10524215 DOI: 10.1016/j.jid.2023.03.1664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/03/2023] [Accepted: 03/02/2023] [Indexed: 03/31/2023]
Abstract
Mouse models that reflect human disorders provide invaluable tools for the translation of basic science discoveries to clinical therapies. However, many of these in vivo therapeutic studies are short term and do not accurately mimic patient conditions. In this study, we used a fully immunocompetent, transgenic mouse model, TGS, in which the spontaneous development of metastatic melanoma is driven by the ectopic expression of a normal neuronal receptor, mGluR1, as a model to assess longitudinal treatment response (up to 8 months) with an inhibitor of glutamatergic signaling, troriluzole, which is a prodrug of riluzole, plus an antibody against PD-1, an immune checkpoint inhibitor. Our results reveal a sex-biased treatment response that led to improved survival in troriluzole and/or anti-PD-1-treated male mice that correlated with differential CD8+ T cells and CD11b+ myeloid cell populations in the tumor-stromal interface, supporting the notion that this model is a responsive and tractable system for evaluating therapeutic regimens for melanoma in an immunocompetent setting.
Collapse
Affiliation(s)
- Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; Graduate Program in Cellular and Molecular Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Kajal Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Allison L Isola
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; ExxonMobil Biomedical Sciences, Annandale, New Jersey, USA
| | - Christina Marinaro
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Maryam Abdur Rasheed
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Joseph Campagnolo
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Mohamad Naser Eddin
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Marco Rossi
- Division of Hematology, Oncology, and Stem Cell Transplant Research, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; Graduate Program in Cellular and Molecular Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Kenneth Reuhl
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA; Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey, USA
| | - Raj Shah
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey, USA
| | - Ann K Robinson
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Anna Chaly
- Fox Chase Therapeutics Discovery, Doylestown, Pennsylvania, USA
| | - Katie B Freeman
- Fox Chase Therapeutics Discovery, Doylestown, Pennsylvania, USA
| | - Wenjin Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Jesus Diaz
- Division of Hematology, Oncology, and Stem Cell Transplant Research, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Philip Furmanski
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA; Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey, USA
| | - Ann W Silk
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Allen B Reitz
- Fox Chase Therapeutics Discovery, Doylestown, Pennsylvania, USA
| | - Andrew Zloza
- Division of Hematology, Oncology, and Stem Cell Transplant Research, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA; Graduate Program in Cellular and Molecular Pharmacology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA; Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, New Jersey, USA.
| |
Collapse
|
2
|
MicroRNA-138-5p Targets Pro-Apoptotic Factors and Favors Neural Cell Survival: Analysis in the Injured Spinal Cord. Biomedicines 2022; 10:biomedicines10071559. [PMID: 35884864 PMCID: PMC9312482 DOI: 10.3390/biomedicines10071559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
The central nervous system microRNA miR-138-5p has attracted much attention in cancer research because it inhibits pro-apoptotic genes including CASP3. We hypothesize that miR-138-5p downregulation after SCI leads to overexpression of pro-apoptotic genes, sensitizing neural cells to noxious stimuli. This study aimed to identify miR-138-5p targets among pro-apoptotic genes overexpressed following SCI and to confirm that miR-138-5p modulates cell death in neural cells. Gene expression and histological analyses revealed that the drop in miR-138-5p expression after SCI is due to the massive loss of neurons and oligodendrocytes and its downregulation in neurons. Computational analyses identified 176 potential targets of miR-138-5p becoming dysregulated after SCI, including apoptotic proteins CASP-3 and CASP-7, and BAK. Reporter, RT-qPCR, and immunoblot assays in neural cell cultures confirmed that miR-138-5p targets their 3′UTRs, reduces their expression and the enzymatic activity of CASP-3 and CASP-7, and protects cells from apoptotic stimuli. Subsequent RT-qPCR and histological analyses in a rat model of SCI revealed that miR-138-5p downregulation correlates with the overexpression of its pro-apoptotic targets. Our results suggest that the downregulation of miR-138-5p after SCI may have deleterious effects on neural cells, particularly on spinal neurons.
Collapse
|
3
|
Razavi SM, Khayatan D, Arab ZN, Momtaz S, Zare K, Jafari RM, Dehpour AR, Abdolghaffari AH. Licofelone, a potent COX/5-LOX inhibitor and a novel option for treatment of neurological disorders. Prostaglandins Other Lipid Mediat 2021; 157:106587. [PMID: 34517113 DOI: 10.1016/j.prostaglandins.2021.106587] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/17/2021] [Accepted: 09/04/2021] [Indexed: 12/13/2022]
Abstract
Neurological disorders result in disability and morbidity. Neuroinflammation is a key factor involved in progression or resolution of a series of neurological disorders like Huntington disease (HD), Parkinson's disease (PD), Alzheimer's disease (AD), Spinal Cord Injury (SCI), and Seizure. Thereby, anti-inflammatory drugs have been developed to improve the neurodegenerative impairments. Licofelone is an approved osteoarthritis drug that inhibits both the COX (cyclooxygenase) and 5-LOX (lipoxygenase) pathways. Licofelone has pain-relieving and anti-inflammatory effects and it was shown to have neuroprotective properties in the central nervous system, which is implicated in its regulatory effect on the COX/5-LOX pathway, inflammatory cytokines, and immune responses. In this study, we briefly review the various features of neurological disorders and the function of COX/LOX in their flare up and current pharmacological products for their management. Moreover, this review attempts to summarize potential therapeutics that target the immune responses within the central nervous system. A better understanding of the interactions between Licofelone and the nervous systems will be crucial to demonstrate the possible efficacy of Licofelone in neurological disorders.
Collapse
Affiliation(s)
- Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran; Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kimia Zare
- School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran; Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
4
|
Nguyen A, Chow DSL, Wu L, Teng YA, Sarkar M, Toups EG, Harrop JS, Schmitt KM, Johnson MM, Guest JD, Aarabi B, Shaffrey CI, Boakye M, Frankowski RF, Fehlings MG, Grossman RG. Longitudinal Impact of Acute Spinal Cord Injury on Clinical Pharmacokinetics of Riluzole, a Potential Neuroprotective Agent. J Clin Pharmacol 2021; 61:1232-1242. [PMID: 33908635 PMCID: PMC8457124 DOI: 10.1002/jcph.1876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/18/2021] [Indexed: 11/09/2022]
Abstract
Riluzole, a benzothiazole sodium channel blocker that received US Food and Drug Administration approval to attenuate neurodegeneration in amyotrophic lateral sclerosis in 1995, was found to be safe and potentially efficacious in a spinal cord injury (SCI) population, as evident in a phase I clinical trial. The acute and progressive nature of traumatic SCI and the complexity of secondary injury processes can alter the pharmacokinetics of therapeutics. A 1-compartment with first-order elimination population pharmacokinetic model for riluzole incorporating time-dependent clearance and volume of distribution was developed from combined data of the phase 1 and the ongoing phase 2/3 trials. This change in therapeutic exposure may lead to a biased estimate of the exposure-response relationship when evaluating therapeutic effects. With the developed model, a rational, optimal dosing scheme can be designed with time-dependent modification that preserves the required therapeutic exposure of riluzole.
Collapse
Affiliation(s)
- Ashley Nguyen
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Diana S-L Chow
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Lei Wu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Yang Angela Teng
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA.,Covance, Madison, Wisconsin, USA
| | - Mahua Sarkar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Elizabeth G Toups
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, Texas, USA
| | - James S Harrop
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Karl M Schmitt
- Department of Neurosurgery, Health Science Center, University of Texas, Houston, Texas, USA
| | - Michele M Johnson
- Department of Neurosurgery, Health Science Center, University of Texas, Houston, Texas, USA.,Atlanta Brain and Spine Care, Atlanta, Georgia, USA
| | - James D Guest
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Bizhan Aarabi
- Department of Neurosurgery, University of Maryland, Baltimore, Maryland, USA
| | - Christopher I Shaffrey
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, Virginia, USA.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Maxwell Boakye
- Department of Neurosurgery, University of Louisville, Louisville, Kentucky, USA
| | - Ralph F Frankowski
- Late colleague, Division of Biostatistics, University of Texas School of Public Health, Houston, Texas, USA
| | - Michael G Fehlings
- Division of Neurosurgery and Spine Program, Toronto Western Hospital, University of, Toronto, Ontario, Canada
| | - Robert G Grossman
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
5
|
MicroRNA-145-Mediated KDM6A Downregulation Enhances Neural Repair after Spinal Cord Injury via the NOTCH2/Abcb1a Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2580619. [PMID: 34122720 PMCID: PMC8169274 DOI: 10.1155/2021/2580619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 01/18/2021] [Accepted: 04/02/2021] [Indexed: 01/11/2023]
Abstract
Spinal cord injury (SCI) causes a significant physical, emotional, social, and economic burden to millions of people. MicroRNAs are known players in the regulatory circuitry of the neural repair in SCI. However, most microRNAs remain uncharacterized. Here, we demonstrate the neuroprotection of microRNA-145 (miR-145) after SCI in vivo and in vitro. In silico analysis predicted the target gene KDM6A of miR-145. The rat SCI model was developed by weight drop, and lipopolysaccharide- (LPS-) induced PC12 cell inflammatory injury model was also established. We manipulated the expression of miR-145 and/or KDM6A both in vivo and in vitro to explain their roles in rat neurological functional recovery as well as PC12 cell activities and inflammation. Furthermore, we delineated the mechanistic involvement of NOTCH2 and Abcb1a in the neuroprotection of miR-145. According to the results, miR-145 was poorly expressed and KDM6A was highly expressed in the spinal cord tissue of the SCI rat model and LPS-induced PC12 cells. Overexpression of miR-145 protects PC12 cells from LPS-induced cell damage and expedites neurological functional recovery of SCI in rats. miR-145 was validated to target and downregulate the demethylase KDM6A expression, thus abrogating the expression of Abcb1a by promoting the methylation of NOTCH2. Additionally, in vivo findings verified that miR-145 expedites neuroprotection after SCI by regulating the KDM6A/NOTCH2/Abcb1a axis. Taken together, miR-145 confers neuroprotective effects and enhances neural repair after SCI through the KDM6A-mediated NOTCH2/Abcb1a axis.
Collapse
|
6
|
Vita SM, Redell JB, Maynard ME, Zhao J, Grill RJ, Dash PK, Grayson BE. P-glycoprotein Expression Is Upregulated in a Pre-Clinical Model of Traumatic Brain Injury. Neurotrauma Rep 2020; 1:207-217. [PMID: 33274346 PMCID: PMC7703495 DOI: 10.1089/neur.2020.0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Athletes participating in contact sports are at risk for sustaining repeat mild traumatic brain injury (rmTBI). Unfortunately, no pharmacological treatment to lessen the pathophysiology of brain injury has received U.S. Food and Drug Administration (FDA) approval. One hurdle to overcome for potential candidate agents to reach effective therapeutic concentrations in the brain is the blood-brain barrier (BBB). Adenosine triphosphate (ATP)-binding cassette (ABC) transporters, such as P-glycoprotein (Pgp), line the luminal membrane of the brain capillary endothelium facing the vascular space. Although these transporters serve to protect the central nervous system (CNS) from damage by effluxing neurotoxicants before they can reach the brain, they may also limit the accumulation of therapeutic drugs in the brain parenchyma. Thus, increased Pgp expression following brain injury may result in reduced brain availability of therapeutic agents. We therefore questioned if repeat concussive injury increases Pgp expression in the brain. To answer this question, we used a rodent model of repeat mild closed head injury (rmCHI) and examined the messenger RNA (mRN) and protein expression of both isoforms of rodent Pgp (Abcb1a and Abcb1b). Compared with sham-operated controls (n = 5), the mRNA levels of both Abcb1a and Abcb1b were found to be increased in the hippocampus at day 1 (n = 5) and at day 5 (n = 5) post-injury. Using a validated antibody, we show increased immunolabeling for Pgp in the dorsal cortex at day 5 and in the hippocampus at day 1 (n = 5) and at day 5 (n = 5) post-injury compared with sham controls (n = 6). Taken together, these results suggest that increased expression of Pgp after rmCHI may reduce the brain accumulation of therapeutic drugs that are Pgp substrates. It is plausible that including a Pgp inhibitor with a candidate therapeutic agent may be an effective approach to treat the pathophysiology of rmCHI.
Collapse
Affiliation(s)
- Sydney M Vita
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Mark E Maynard
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Raymond J Grill
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Bernadette E Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
7
|
Chiang M, Back HM, Lee JB, Oh S, Guo T, Girgis S, Park C, Haroutounian S, Kagan L. Pharmacokinetic Modeling of the Impact of P-glycoprotein on Ondansetron Disposition in the Central Nervous System. Pharm Res 2020; 37:205. [PMID: 32989520 DOI: 10.1007/s11095-020-02929-2] [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: 04/26/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Modulation of 5-HT3 receptor in the central nervous system (CNS) is a promising approach for treatment of neuropathic pain. The goal was to evaluate the role of P-glycoprotein (Pgp) in limiting exposure of different parts of the CNS to ondansetron (5-HT3 receptor antagonist) using wild-type and genetic knockout rat model. METHODS Plasma pharmacokinetics and CNS (brain, spinal cord, and cerebrospinal fluid) disposition was studied after single 10 mg/kg intravenous dose. RESULTS Pgp knockout resulted in significantly higher concentrations of ondansetron in all tested regions of the CNS at most of the time points. The mean ratio of the concentrations between KO and WT animals was 2.39-5.48, depending on the region of the CNS. Male and female animals demonstrated some difference in ondansetron plasma pharmacokinetics and CNS disposition. Mechanistic pharmacokinetic model that included two systemic disposition and three CNS compartments (with intercompartmental exchange) was developed. Pgp transport was incorporated as an efflux from the brain and spinal cord to the central compartment. The model provided good simultaneous description of all data sets, and all parameters were estimated with sufficient precision. CONCLUSIONS The study provides important quantitative information on the role of Pgp in limiting ondansetron exposure in various regions of the CNS using data from wild-type and Pgp knockout rats. CSF drug concentrations, as a surrogate to CNS exposure, are likely to underestimate the effect of Pgp on drug penetration to the brain and the spinal cord.
Collapse
Affiliation(s)
- Manting Chiang
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA.,Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hyun-Moon Back
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA.,Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Jong Bong Lee
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | - Sarah Oh
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | - Tiffany Guo
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | - Simone Girgis
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | - Celine Park
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA
| | - Simon Haroutounian
- Division of Clinical and Translational Research and Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario, School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, New Jersey, 08854, USA. .,Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA.
| |
Collapse
|
8
|
van Vliet EA, Iyer AM, Mesarosova L, Çolakoglu H, Anink JJ, van Tellingen O, Maragakis NJ, Shefner J, Bunt T, Aronica E. Expression and Cellular Distribution of P-Glycoprotein and Breast Cancer Resistance Protein in Amyotrophic Lateral Sclerosis Patients. J Neuropathol Exp Neurol 2020; 79:266-276. [PMID: 31999342 PMCID: PMC7036662 DOI: 10.1093/jnen/nlz142] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 11/23/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
For amyotrophic lateral sclerosis (ALS), achieving and maintaining effective drug levels in the brain is challenging due to the activity of ATP-binding cassette (ABC) transporters which efflux drugs that affect drug exposure and response in the brain. We investigated the expression and cellular distribution of the ABC transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) using immunohistochemistry in spinal cord (SC), motor cortex, and cerebellum from a large cohort of genetically well characterized ALS patients (n = 25) and controls (n = 14). The ALS group included 17 sporadic (sALS) and 8 familial (fALS) patients. Strong P-gp expression was observed in endothelial cells in both control and ALS specimens. Immunohistochemical analysis showed higher P-gp expression in reactive astroglial cells in both gray (ventral horn) and white matter of the SC, as well as in the motor cortex of all ALS patients, as compared with controls. BCRP expression was higher in glia in the SC and in blood vessels and glia in the motor cortex of ALS patients, as compared with controls. P-gp and BCRP immunoreactivity did not differ between sALS and fALS cases. The upregulation of both ABC transporters in the brain may explain multidrug resistance in ALS patients and has implications for the use of both approved and experimental therapeutics.
Collapse
Affiliation(s)
- Erwin A van Vliet
- From the Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam
| | - Anand M Iyer
- From the Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience
| | - Lucia Mesarosova
- From the Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience
| | - Hilal Çolakoglu
- Division of Pharmacology, The Netherlands Cancer Institute (HÇ, OvT), Amsterdam, The Netherlands
| | - Jasper J Anink
- From the Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute (HÇ, OvT), Amsterdam, The Netherlands
| | - Nicholas J Maragakis
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeremy Shefner
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, Massachusetts
| | - Eleonora Aronica
- From the Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience
| |
Collapse
|
9
|
Srinivas S, Wali AR, Pham MH. Efficacy of riluzole in the treatment of spinal cord injury: a systematic review of the literature. Neurosurg Focus 2020; 46:E6. [PMID: 30835675 DOI: 10.3171/2019.1.focus18596] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/02/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVERiluzole is a glutamatergic modulator that has recently shown potential for neuroprotection after spinal cord injury (SCI). While the effects of riluzole are extensively documented in animal models of SCI, there remains heterogeneity in findings. Moreover, there is a paucity of data on the pharmacology of riluzole and its effects in humans. For the present study, the authors systematically reviewed the literature to provide a comprehensive understanding of the effects of riluzole in SCI.METHODSThe PubMed database was queried from 1996 to September 2018 to identify animal studies and clinical trials involving riluzole administration for SCI. Once articles were identified, they were processed for year of publication, study design, subject type, injury model, number of subjects in experimental and control groups, dose, timing/route of administration, and outcomes.RESULTSA total of 37 studies were included in this study. Three placebo-controlled clinical trials were included with a total of 73 patients with a mean age of 39.1 years (range 18-70 years). For the clinical trials included within this study, the American Spinal Injury Association Impairment Scale distributions for SCI were 42.6% grade A, 25% grade B, 26.6% grade C, and 6.2% grade D. Key findings from studies in humans included decreased nociception, improved motor function, and attenuated spastic reflexes. Twenty-six animal studies (24 in vivo, 1 in vitro, and 1 including both in vivo and in vitro) were included. A total of 520 animals/in vitro specimens were exposed to riluzole and 515 animals/in vitro specimens underwent other treatment for comparison. The average dose of riluzole for intraperitoneal, in vivo studies was 6.5 mg/kg (range 1-10 mg/kg). Key findings from animal studies included behavioral improvement, histopathological tissue sparing, and modified electrophysiology after SCI. Eight studies examined the pharmacology of riluzole in SCI. Key findings from pharmacological studies included riluzole dose-dependent effects on glutamate uptake and its modified bioavailability after SCI in both animal and clinical models.CONCLUSIONSSCI has many negative sequelae requiring neuroprotective intervention. While still relatively new in its applications for SCI, both animal and human studies demonstrate riluzole to be a promising pharmacological intervention to attenuate the devastating effects of this condition.
Collapse
|
10
|
Koehn LM. ABC efflux transporters at blood-central nervous system barriers and their implications for treating spinal cord disorders. Neural Regen Res 2020; 15:1235-1242. [PMID: 31960802 PMCID: PMC7047801 DOI: 10.4103/1673-5374.272568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The barriers present in the interfaces between the blood and the central nervous system form a major hurdle for the pharmacological treatment of central nervous system injuries and diseases. The family of ATP-binding cassette (ABC) transporters has been widely studied regarding efflux of medications at blood-central nervous system barriers. These efflux transporters include P-glycoprotein (abcb1), 'breast cancer resistance protein' (abcg2) and the various 'multidrug resistance-associated proteins' (abccs). Understanding which efflux transporters are present at the blood-spinal cord, blood-cerebrospinal fluid and cerebrospinal fluid-spinal cord barriers is necessary to determine their involvement in limiting drug transfer from blood to the spinal cord tissue. Recent developments in the blood-brain barrier field have shown that barrier systems are dynamic and the profile of barrier defenses can alter due to conditions such as age, disease and environmental challenge. This means that a true understanding of ABC efflux transporter expression and localization should not be one static value but instead a range that represents the complex patient subpopulations that exist. In the present review, the blood-central nervous system barrier literature is discussed with a focus on the impact of ABC efflux transporters on: (i) protecting the spinal cord from adverse effects of systemically directed drugs, and (ii) limiting centrally directed drugs from accessing their active sites within the spinal cord.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pharmacology and Therapeutics, the University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
11
|
Zhou LY, Tian ZR, Yao M, Chen XQ, Song YJ, Ye J, Yi NX, Cui XJ, Wang YJ. Riluzole promotes neurological function recovery and inhibits damage extension in rats following spinal cord injury: a meta-analysis and systematic review. J Neurochem 2019; 150:6-27. [PMID: 30786027 DOI: 10.1111/jnc.14686] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/03/2019] [Accepted: 02/15/2019] [Indexed: 12/24/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition that has few treatment options. Riluzole, a sodium channel blocker used to treat amyotrophic lateral sclerosis, has been initially trialed in human SCI. We performed a systematic review to critically assess the efficacy of riluzole in locomotor recovery and damage extension in SCI rat models, and the potential for clinical translation. PubMed, Embase, Cochrane Library, and Chinese databases were searched from their inception date to March 2018. Two reviewers independently selected animal studies that evaluated neurological recovery and lesion area following riluzole treatment in SCI rat models, extracted data and assessed methodological quality. Pairwise meta-analysis, subgroup analysis, and network meta-analysis were performed to assess the effects of riluzole on SCI. Ten eligible studies were included. Two studies had high methodological quality. Overall, the Basso, Beattie, and Bresnahan scores were increased in riluzole-treated animals versus controls, and effect sizes showed a gradual increase from the 1st (five studies, n = 104, mean difference = 1.24, 95% CI = 0.11 to 2.37, p = 0.03) to 6th week after treatment (five studies, n = 120, mean difference = 2.34, 95% CI = 1.26 to 3.42, p < 0.0001). Riluzole was associated with improved outcomes in the inclined plane test and the tissue preservation area. Subgroup analyses suggested an association of locomotor recovery with riluzole dose. Network meta-analysis showed that 5 mg/kg riluzole exhibited greater protection than 2.5 and 8 mg/kg riluzole. Collectively, this review suggests that riluzole has a protective effect on SCI, with good safety and a clear mechanism of action and may be suitable for future clinical trials or applications. However, animal results should be interpreted with caution given the known limitations in animal experimental design and methodological quality.
Collapse
Affiliation(s)
- Long-Yun Zhou
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Rehabilitation Medicine College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zi-Rui Tian
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu-Qing Chen
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yong-Jia Song
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ye
- Department of Orthopedics and Traumatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nan-Xing Yi
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue-Jun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Jun Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Kar S, Ramamoorthy G, Sinha S, Ramanan M, Pola JK, Golakoti NR, Nanubolu JB, Sahoo SK, Dandamudi RB, Doble M. Synthesis of diarylidenecyclohexanone derivatives as potential anti-inflammatory leads against COX-2/mPGES1 and 5-LOX. NEW J CHEM 2019. [DOI: 10.1039/c9nj00726a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study establishes the diarylidenecyclohexanones as good anti-inflammatory pharmacophores with selective high potency against PGE2and 5-LOX without toxicity towards healthy human cells.
Collapse
Affiliation(s)
- Swayamsiddha Kar
- Department of Chemistry
- Sri Sathya Sai Institute of Higher Learning
- India
| | - Gayathri Ramamoorthy
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - Shweta Sinha
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - Meera Ramanan
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - Jeevan Kumar Pola
- Department of Chemistry
- Sri Sathya Sai Institute of Higher Learning
- India
| | | | | | - Suraj Kumar Sahoo
- Department of Chemistry
- Sri Sathya Sai Institute of Higher Learning
- India
| | | | - Mukesh Doble
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| |
Collapse
|
13
|
Devi NS, Ramanan M, Paragi-Vedanthi P, Doble M. Phytochemicals as multi-target inhibitors of the inflammatory pathway- A modeling and experimental study. Biochem Biophys Res Commun 2017; 484:467-473. [DOI: 10.1016/j.bbrc.2017.01.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 02/01/2023]
|
14
|
Hook MA, Woller SA, Bancroft E, Aceves M, Funk MK, Hartman J, Garraway SM. Neurobiological Effects of Morphine after Spinal Cord Injury. J Neurotrauma 2016; 34:632-644. [PMID: 27762659 DOI: 10.1089/neu.2016.4507] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Opioids and non-steroidal anti-inflammatory drugs are used commonly to manage pain in the early phase of spinal cord injury (SCI). Despite its analgesic efficacy, however, our studies suggest that intrathecal morphine undermines locomotor recovery and increases lesion size in a rodent model of SCI. Similarly, intravenous (IV) morphine attenuates locomotor recovery. The current study explores whether IV morphine also increases lesion size after a spinal contusion (T12) injury and quantifies the cell types that are affected by early opioid administration. Using an experimenter-administered escalating dose of IV morphine across the first seven days post-injury, we quantified the expression of neuron, astrocyte, and microglial markers at the injury site. SCI decreased NeuN expression relative to shams. In subjects with SCI treated with IV morphine, virtually no NeuN+ cells remained across the rostral-caudal extent of the lesion. Further, whereas SCI per se increased the expression of astrocyte and microglial markers (glial fibrillary acidic protein and OX-42, respectively), morphine treatment decreased the expression of these markers. These cellular changes were accompanied by attenuation of locomotor recovery (Basso, Beattie, Bresnahan scores), decreased weight gain, and the development of opioid-induced hyperalgesia (increased tactile reactivity) in morphine-treated subjects. These data suggest that morphine use is contraindicated in the acute phase of a spinal injury. Faced with a lifetime of intractable pain, however, simply removing any effective analgesic for the management of SCI pain is not an ideal option. Instead, these data underscore the critical need for further understanding of the molecular pathways engaged by conventional medications within the pathophysiological context of an injury.
Collapse
Affiliation(s)
- Michelle A Hook
- 1 Texas A&M University Institute for Neuroscience, Texas A&M University , College Station, Texas.,2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Sarah A Woller
- 3 Department of Anesthesiology, University of California , San Diego, California
| | - Eric Bancroft
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Miriam Aceves
- 1 Texas A&M University Institute for Neuroscience, Texas A&M University , College Station, Texas.,2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Mary Katherine Funk
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - John Hartman
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Sandra M Garraway
- 4 Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
15
|
Kabu S, Gao Y, Kwon BK, Labhasetwar V. Drug delivery, cell-based therapies, and tissue engineering approaches for spinal cord injury. J Control Release 2015; 219:141-154. [PMID: 26343846 DOI: 10.1016/j.jconrel.2015.08.060] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/23/2015] [Accepted: 08/31/2015] [Indexed: 12/28/2022]
Abstract
Spinal cord injury (SCI) results in devastating neurological and pathological consequences, causing major dysfunction to the motor, sensory, and autonomic systems. The primary traumatic injury to the spinal cord triggers a cascade of acute and chronic degenerative events, leading to further secondary injury. Many therapeutic strategies have been developed to potentially intervene in these progressive neurodegenerative events and minimize secondary damage to the spinal cord. Additionally, significant efforts have been directed toward regenerative therapies that may facilitate neuronal repair and establish connectivity across the injury site. Despite the promise that these approaches have shown in preclinical animal models of SCI, challenges with respect to successful clinical translation still remain. The factors that could have contributed to failure include important biologic and physiologic differences between the preclinical models and the human condition, study designs that do not mirror clinical reality, discrepancies in dosing and the timing of therapeutic interventions, and dose-limiting toxicity. With a better understanding of the pathobiology of events following acute SCI, developing integrated approaches aimed at preventing secondary damage and also facilitating neuroregenerative recovery is possible and hopefully will lead to effective treatments for this devastating injury. The focus of this review is to highlight the progress that has been made in drug therapies and delivery systems, and also cell-based and tissue engineering approaches for SCI.
Collapse
Affiliation(s)
- Shushi Kabu
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yue Gao
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Brian K Kwon
- Department of Orthopaedics, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada V5Z 1M9
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
16
|
Dennys CN, Armstrong J, Levy M, Byun YJ, Ramdial KR, Bott M, Rossi FH, Fernández-Valle C, Franco MC, Estevez AG. Chronic inhibitory effect of riluzole on trophic factor production. Exp Neurol 2015; 271:301-7. [PMID: 26071088 PMCID: PMC4864959 DOI: 10.1016/j.expneurol.2015.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/18/2015] [Accepted: 05/23/2015] [Indexed: 12/14/2022]
Abstract
Riluzole is the only FDA approved drug for the treatment of amyotrophic lateral sclerosis (ALS). However, the drug affords moderate protection to ALS patients, extending life for a few months by a mechanism that remains controversial. In the presence of riluzole, astrocytes increase the production of factors protective to motor neurons. The stimulation of trophic factor production by motor neuron associated cells may contribute to riluzole's protective effect in ALS. Here, we investigated the effects of media conditioned by astrocytes and Schwann cells acutely or chronically incubated with riluzole on trophic factor-deprived motor neuron survival. While acute riluzole incubation induced CT-1 secretion by astrocytes and Schwann cells, chronic treatment stimulated a significant decrease in trophic factor production compared to untreated cultures. Accordingly, conditioned media from astrocytes and Schwann cells acutely treated with riluzole protected motor neurons from trophic factor deprivation-induced cell death. Motor neuron protection was prevented by incubation with CT-1 neutralizing antibodies. In contrast, conditioned media from astrocytes and Schwann cells chronically treated with riluzole was not protective. Acute and chronic treatment of mice with riluzole showed opposite effects on trophic factor production in spinal cord, sciatic nerve and brain. There was an increase in the production of CT-1 and GDNF in the spinal cord and CT-1 in the sciatic nerve during the first days of treatment with riluzole, but the levels dropped significantly after chronic treatment with the drug. Similar results were observed in brain for CT-1 and BDNF while there was no change in GDNF levels after riluzole treatment. Our results reveal that riluzole regulates long-lasting processes involving protein synthesis, which may be relevant for riluzole therapeutic effects. Changing the regimen of riluzole administration to favor the acute effect of the drug on trophic factor production by discontinuous long-term treatment may improve the outcome of ALS patient therapy.
Collapse
Affiliation(s)
- Cassandra N Dennys
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - JeNay Armstrong
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Mark Levy
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Youn Jung Byun
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Kristina R Ramdial
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Marga Bott
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Fabian H Rossi
- Orlando Veteran Administration Healthcare System, Orlando, FL 32803, United States
| | - Cristina Fernández-Valle
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Maria Clara Franco
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Alvaro G Estevez
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States.
| |
Collapse
|
17
|
Jablonski M, Miller DS, Pasinelli P, Trotti D. ABC transporter-driven pharmacoresistance in Amyotrophic Lateral Sclerosis. Brain Res 2015; 1607:1-14. [PMID: 25175835 PMCID: PMC4344920 DOI: 10.1016/j.brainres.2014.08.060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/19/2014] [Indexed: 12/12/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a slowly progressing neurodegenerative disease that affects motor neurons of the nervous system. Despite the identification of many potential therapeutics targeting pathogenic mechanisms in in vitro models, there has been limited progress in translating them into a successful pharmacotherapy in the animal model of ALS. Further, efforts to translate any promising results from preclinical trials to effective pharmacotherapies for patients have been unsuccessful, with the exception of riluzole, the only FDA-approved medication, which only modestly extends survival both in the animal model and in patients. Thus, it is essential to reconsider the strategies for developing ALS pharmacotherapies. Growing evidence suggests that problems identifying highly effective ALS treatments may result from an underestimated issue of drug bioavailability and disease-driven pharmacoresistance, mediated by the ATP-binding cassette (ABC) drug efflux transporters. ABC transporters are predominately localized to the lumen of endothelial cells of the blood-brain and blood-spinal cord barriers (BBB, BSCB) where they limit the entry into the central nervous system (CNS) of a wide range of neurotoxicants and xenobiotics, but also therapeutics. In ALS, expression and function of ABC transporters is increased at the BBB/BSCB and their expression has been detected on neurons and glia in the CNS parenchyma, which may further reduce therapeutic action in target cells. Understanding and accounting for the contribution of these transporters to ALS pharmacoresistance could both improve the modest effects of riluzole and set in motion a re-evaluation of previous ALS drug disappointments. In addition, identifying pathogenic mechanisms regulating ABC transporter expression and function in ALS may lead to the development of new therapeutic strategies. It is likely that novel pharmacological approaches require counteracting pharmacoresistance to improve therapeutic efficacy. This article is part of a Special Issue entitled ALS complex pathogenesis.
Collapse
Affiliation(s)
- Michael Jablonski
- Weinberg Unit for ALS Research, Farber Institute for Neurosciences, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19004, USA.
| | - David S Miller
- Laboratory of Toxicology and Pharmacology, NIH/NIEHS, Research Triangle Park, NC 27709, USA
| | - Piera Pasinelli
- Weinberg Unit for ALS Research, Farber Institute for Neurosciences, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19004, USA
| | - Davide Trotti
- Weinberg Unit for ALS Research, Farber Institute for Neurosciences, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19004, USA.
| |
Collapse
|
18
|
Zhang K, Zhao J, Su W, Lu R, Lv P. Immunomodulatory effectiveness of licofelone in preventing epidural fibrosis in post-laminectomy rat. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2014; 25 Suppl 1:S63-8. [PMID: 25200318 DOI: 10.1007/s00590-014-1534-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/03/2014] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The object of this study was to investigate the effects of licofelone on the prevention of epidural fibrosis (EF) formation in post-laminectomy rat models. METHODS A controlled double-blinded study was conducted in sixty, healthy adult Wistar rats that underwent laminectomy at the L1-L2 vertebrae levels. All the rats were divided randomly into three groups according to the treatment (via oral gavage): (1) licofelone treatment group; (2) vehicle treatment group; (3) sham group (laminectomy without treatment). All rats were euthanized humanely 4 weeks postoperatively. The macroscopic assessment of EF, hydroxyproline content in epidural scar tissue, histological analysis, and the mRNA measurements of interleukin-6 (IL-6) and transforming growth factor-β1 were performed. RESULTS The Rydell score, hydroxyproline content, epidural scar density, and inflammatory factors expressions all suggested better results in licofelone group than the other two groups. CONCLUSION The application of licofelone could reduce hydroxyproline deposits, inflammatory factors expressions and prevent epidural adhesions in post-laminectomy rats.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | | | | | | | | |
Collapse
|
19
|
Walters ET. Neuroinflammatory contributions to pain after SCI: roles for central glial mechanisms and nociceptor-mediated host defense. Exp Neurol 2014; 258:48-61. [PMID: 25017887 DOI: 10.1016/j.expneurol.2014.02.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/23/2014] [Accepted: 02/02/2014] [Indexed: 12/30/2022]
Abstract
Neuropathic pain after spinal cord injury (SCI) is common, often intractable, and can be severely debilitating. A number of mechanisms have been proposed for this pain, which are discussed briefly, along with methods for revealing SCI pain in animal models, such as the recently applied conditioned place preference test. During the last decade, studies of animal models have shown that both central neuroinflammation and behavioral hypersensitivity (indirect reflex measures of pain) persist chronically after SCI. Interventions that reduce neuroinflammation have been found to ameliorate pain-related behavior, such as treatment with agents that inhibit the activation states of microglia and/or astroglia (including IL-10, minocycline, etanercept, propentofylline, ibudilast, licofelone, SP600125, carbenoxolone). Reversal of pain-related behavior has also been shown with disruption by an inhibitor (CR8) and/or genetic deletion of cell cycle-related proteins, deletion of a truncated receptor (trkB.T1) for brain-derived neurotrophic factor (BDNF), or reduction by antisense knockdown or an inhibitor (AMG9810) of the activity of channels (TRPV1 or Nav1.8) important for electrical activity in primary nociceptors. Nociceptor activity is known to drive central neuroinflammation in peripheral injury models, and nociceptors appear to be an integral component of host defense. Thus, emerging results suggest that spinal and systemic effects of SCI can activate nociceptor-mediated host defense responses that interact via neuroinflammatory signaling with complex central consequences of SCI to drive chronic pain. This broader view of SCI-induced neuroinflammation suggests new targets, and additional complications, for efforts to develop effective treatments for neuropathic SCI pain.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, TX, USA.
| |
Collapse
|
20
|
Tucker SC, Honn KV. Emerging targets in lipid-based therapy. Biochem Pharmacol 2013; 85:673-688. [PMID: 23261527 PMCID: PMC4106802 DOI: 10.1016/j.bcp.2012.11.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 11/29/2012] [Accepted: 11/30/2012] [Indexed: 02/07/2023]
Abstract
The use of prostaglandins and NSAIDS in the clinic has proven that lipid mediators and their associated pathways make attractive therapeutic targets. When contemplating therapies involving lipid pathways, several basic agents come to mind. There are the enzymes and accessory proteins that lead to the metabolism of lipid substrates, provided through diet or through actions of lipases, the subsequent lipid products, and finally the lipid sensors or receptors. There is abundant evidence that molecules along this lipid continuum can serve as prognostic and diagnostic indicators and are in fact viable therapeutic targets. Furthermore, lipids themselves can be used as therapeutics. Despite this, the vernacular dialog pertaining to "biomarkers" does not routinely include mention of lipids, though this is rapidly changing. Collectively these agents are becoming more appreciated for their respective roles in diverse disease processes from cancer to preterm labor and are receiving their due appreciation after decades of ground work in the lipid field. By relating examples of disease processes that result from dysfunction along the lipid continuum, as well as examples of lipid therapies and emerging technologies, this review is meant to inspire further reading and discovery.
Collapse
Affiliation(s)
- Stephanie C Tucker
- Department of Pathology, Wayne State University School of Medicine, and Karmanos Cancer Institute, Detroit, MI 48202, USA.
| | - Kenneth V Honn
- Department of Pathology, Wayne State University School of Medicine, and Karmanos Cancer Institute, Detroit, MI 48202, USA; Department of Chemistry, Wayne State University School of Medicine, and Karmanos Cancer Institute, Detroit, MI 48202, USA.
| |
Collapse
|
21
|
Dileep KV, Remya C, Tintu I, Haridas M, Sadasivan C. Interactions of selected indole derivatives with phospholipase A₂: in silico and in vitro analysis. J Mol Model 2013; 19:1811-7. [PMID: 23315198 DOI: 10.1007/s00894-012-1741-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/17/2012] [Indexed: 11/30/2022]
Abstract
Phospholipase A2 (PLA₂) is one of the key enzymes involved in the formation of inflammatory mediators. Inhibition of PLA₂ is considered to be one of the efficient methods to control inflammation. In silico docking studies of 160 selected indole derivatives performed against porcine pancreatic PLA₂ (ppsPLA2) suggested that, CID2324681, CID8617 (indolebutyric acid or IBA), CID22097771 and CID802 (indoleacetic acid or IAA) exhibited highest binding energies. In silico analysis was carried out to predict some of the ADME properties. The binding potential of these compounds with human non pancreatic secretory PLA₂ (hnpsPLA₂) was determined using molecular docking studies. In order to corroborate the in silico results, enzyme kinetics and isothermal titration calorimetric analysis of the two selected compounds, IAA and IBA were performed against ppsPLA₂. From the analysis, it was concluded that IAA and IBA can act as competitive inhibitors to the enzyme and may be used as anti inflammatory agents.
Collapse
Affiliation(s)
- Kalarickal Vijayan Dileep
- Department of Biotechnology and Microbiology and Inter-University Centre for Bioscience, Kannur University, Thalassery Campus, Kannur, Palayad 670661, India
| | | | | | | | | |
Collapse
|