1
|
Wang X, Wang Y, Yu N, Xu H, Lei Z. Observational study of ropivacaine and compound betamethasone mixture for analgesia after triangular fibrocartilage complex repair under wrist arthroscopy: A single-center randomized double-blind controlled trial. J Orthop Sci 2024; 29:1208-1213. [PMID: 37863682 DOI: 10.1016/j.jos.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/18/2023] [Accepted: 08/26/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND The purpose of this study was to investigate the clinical effect of an intra-articular and local infiltration injection of a compound analgesic mixture of ropivacaine and compound betamethasone on the repair of the triangular fibrocartilage complex under wrist arthroscopy. METHODS This prospective, double-blind, randomized study involved 20 patients with Atzei type 2 or 3 injuries of the triangular fibrocartilage complex who underwent repair under wrist arthroscopy. Patients were divided into two groups (n = 10) according to the systematic random sampling method. The test group was injected with a "cocktail" mixture for pain relief. The control group was injected with normal saline. The visual analog scale (VAS) pain score, pinch force, wrist joint mobility, wrist joint function score (PRWE score), occurrence of adverse reactions and dosage of analgesic drugs were evaluated before and after the operation in the two groups. RESULTS The resting pain of the patients in the test group was less severe than that of the control group at 12 h, 24 h and 48 h after the operation (P < 0.05), and the pinch force of the patients in the test group was significantly greater than that of the control group at 1 d, 2 d and 3 d after the operation (P < 0.01). The amount of postoperative analgesics used in the test group was significantly lower than that in the control group (P < 0.01), and the patient satisfaction rate in the test group was higher than that in the control group (P < 0.05). There were no postoperative adverse effects in either group. CONCLUSION An intra-articular and local infiltration injection of a "cocktail" analgesic mixture in the repair of triangular fibrocartilage complex under wrist arthroscopy can provide good pain control in the early postoperative period and reduce the amount of postoperative analgesic drugs administered, thus improving clinical safety. LEVEL OF EVIDENCE Level II; Randomized Controlled Trial; Treatment Study.
Collapse
Affiliation(s)
- Xinzhu Wang
- Hand Surgery 5 Ward, Central Hospital Affiliated to Shenyang Medical College. No. 5, Nanqi West Road, Tiexi District, Shenyang City, Liaoning Province, China
| | - Yansheng Wang
- Hand Surgery 5 Ward, Central Hospital Affiliated to Shenyang Medical College. No. 5, Nanqi West Road, Tiexi District, Shenyang City, Liaoning Province, China
| | - Ning Yu
- Hand Surgery 5 Ward, Central Hospital Affiliated to Shenyang Medical College. No. 5, Nanqi West Road, Tiexi District, Shenyang City, Liaoning Province, China
| | - Hui Xu
- Hand Surgery 5 Ward, Central Hospital Affiliated to Shenyang Medical College. No. 5, Nanqi West Road, Tiexi District, Shenyang City, Liaoning Province, China
| | - Zeming Lei
- Hand Surgery 5 Ward, Central Hospital Affiliated to Shenyang Medical College. No. 5, Nanqi West Road, Tiexi District, Shenyang City, Liaoning Province, China; Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University. No. 36, Sanhao Street, Heping District, Shenyang City, Liaoning Province, China.
| |
Collapse
|
2
|
Sudhahar S, Ozer B, Chang J, Chadwick W, O'Donovan D, Campbell A, Tulip E, Thompson N, Roberts I. An experimentally validated approach to automated biological evidence generation in drug discovery using knowledge graphs. Nat Commun 2024; 15:5703. [PMID: 38977662 PMCID: PMC11231212 DOI: 10.1038/s41467-024-50024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Explaining predictions for drug repositioning with biological knowledge graphs is a challenging problem. Graph completion methods using symbolic reasoning predict drug treatments and associated rules to generate evidence representing the therapeutic basis of the drug. Yet the vast amounts of generated paths that are biologically irrelevant or not mechanistically meaningful within the context of disease biology can limit utility. We use a reinforcement learning based knowledge graph completion model combined with an automatic filtering approach that produces the most relevant rules and biological paths explaining the predicted drug's therapeutic connection to the disease. In this work we validate the approach against preclinical experimental data for Fragile X syndrome demonstrating strong correlation between automatically extracted paths and experimentally derived transcriptional changes of selected genes and pathways of drug predictions Sulindac and Ibudilast. Additionally, we show it reduces the number of generated paths in two case studies, 85% for Cystic fibrosis and 95% for Parkinson's disease.
Collapse
|
3
|
Grodin EN. Neuroimmune modulators as novel pharmacotherapies for substance use disorders. Brain Behav Immun Health 2024; 36:100744. [PMID: 38435721 PMCID: PMC10906159 DOI: 10.1016/j.bbih.2024.100744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
One promising avenue of research is the use of neuroimmune modulators to treat substance use disorders (SUDs). Neuroimmune modulators target the interactions between the nervous system and immune system, which have been found to play a crucial role in the development and maintenance of SUDs. Multiple classes of substances produce alterations to neuroimmune signaling and peripheral immune function, including alcohol, opioids, and psychostimulants Preclinical studies have shown that neuroimmune modulators can reduce drug-seeking behavior and prevent relapse in animal models of SUDs. Additionally, early-phase clinical trials have demonstrated the safety and feasibility of using neuroimmune modulators as a treatment for SUDs in humans. These therapeutics can be used as stand-alone treatments or as adjunctive. This review summarizes the current state of the field and provides future directions with a specific focus on personalized medicine.
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
- Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
4
|
Chadwick W, Angulo-Herrera I, Cogram P, Deacon RJM, Mason DJ, Brown D, Roberts I, O’Donovan DJ, Tranfaglia MR, Guilliams T, Thompson NT. A novel combination treatment for fragile X syndrome predicted using computational methods. Brain Commun 2024; 6:fcad353. [PMID: 38226317 PMCID: PMC10789243 DOI: 10.1093/braincomms/fcad353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/07/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Fragile X syndrome is a neurodevelopmental disorder caused by silencing of the fragile X messenger ribonucleotide gene. Patients display a wide spectrum of symptoms ranging from intellectual and learning disabilities to behavioural challenges including autism spectrum disorder. In addition to this, patients also display a diversity of symptoms due to mosaicism. These factors make fragile X syndrome a difficult syndrome to manage and suggest that a single targeted therapeutic approach cannot address all the symptoms. To this end, we utilized Healx's data-driven drug discovery platform to identify a treatment strategy to address the wide range of diverse symptoms among patients. Computational methods identified the combination of ibudilast and gaboxadol as a treatment for several pathophysiological targets that could potentially reverse multiple symptoms associated with fragile X syndrome. Ibudilast is an approved broad-spectrum phosphodiesterase inhibitor, selective against both phosphodiesterase 4 and phosphodiesterase 10, and has demonstrated to have several beneficial effects in the brain. Gaboxadol is a GABAA receptor agonist, selective against the delta subunit, which has previously displayed encouraging results in a fragile X syndrome clinical trial. Alterations in GABA and cyclic adenosine monophosphate metabolism have long since been associated with the pathophysiology of fragile X syndrome; however, targeting both pathways simultaneously has never been investigated. Both drugs have a good safety and tolerability profile in the clinic making them attractive candidates for repurposing. We set out to explore whether the combination of ibudilast and gaboxadol could demonstrate therapeutic efficacy in a fragile X syndrome mouse model. We found that daily treatment with ibudilast significantly enhanced the ability of fragile X syndrome mice to perform a number of different cognitive assays while gaboxadol treatment improved behaviours such as hyperactivity, aggression, stereotypy and anxiety. Importantly, when ibudilast and gaboxadol were co-administered, the cognitive deficits as well as the aforementioned behaviours were rescued. Moreover, this combination treatment showed no evidence of tolerance, and no adverse effects were reported following chronic dosing. This work demonstrates for the first time that by targeting multiple pathways, with a combination treatment, we were able to rescue more phenotypes in a fragile X syndrome mouse model than either ibudilast or gaboxadol could achieve as monotherapies. This combination treatment approach holds promise for addressing the wide spectrum of diverse symptoms in this heterogeneous patient population and may have therapeutic potential for idiopathic autism.
Collapse
Affiliation(s)
| | | | - Patricia Cogram
- Department of Genetics, Faculty of Science, Institute of Ecology and Biodiversity (IEB), University of Chile, Santiago 7800024, Chile
- Center for Neural Circuit Mapping, UCI, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Robert J M Deacon
- Department of Genetics, Faculty of Science, Institute of Ecology and Biodiversity (IEB), University of Chile, Santiago 7800024, Chile
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Sandhu A, Rawat K, Gautam V, Sharma A, Kumar A, Saha L. Phosphodiesterase inhibitor, ibudilast alleviates core behavioral and biochemical deficits in the prenatal valproic acid exposure model of autism spectrum disorder. Brain Res 2023; 1815:148443. [PMID: 37290608 DOI: 10.1016/j.brainres.2023.148443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/12/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is categorized as a neurodevelopmental disorder, presenting with a variety of aetiological and phenotypical features. Ibudilast is known to produce beneficial effects in several neurological disorders including neuropathic pain, multiple sclerosis, etc. by displaying its neuroprotective and anti-inflammatory properties. Here, in our study, the pharmacological outcome of ibudilast administration was investigated in the prenatal valproic acid (VPA)-model of ASD in Wistar rats. METHODS Autistic-like symptoms were induced in Wistar male pups of dams administered with Valproic acid (VPA) on embryonic day 12.5. VPA-exposed male pups were administered with two doses of ibudilast (5 and10 mg/kg) and all the groups were evaluated for behavioral parameters like social interaction, spatial memory/learning, anxiety, locomotor activity, and nociceptive threshold. Further, the possible neuroprotective effect of ibudilast was evaluated by assessing oxidative stress, neuroinflammation (IL-1β, TNF-α, IL-6, IL-10) in the hippocampus, % area of Glial fibrillary acidic protein (GFAP)-positive cells and neuronal damage in the cerebellum. KEY FINDINGS Treatment with ibudilast significantly attenuated prenatal VPA exposure associated social interaction and spatial learning/memory deficits, anxiety, hyperactivity, and increased nociceptive threshold, and it decreased oxidative stress markers, pro-inflammatory markers (IL-1β, TNF-α, IL-6), and % area of GFAP-positive cells and restored neuronal damage. CONCLUSIONS Ibudilast treatment has restored crucial ASD-related behavioural abnormalities, potentially through neuroprotection. Therefore, benefits of ibudilast administration in animal models of ASD suggest that ibudilast may have therapeutic potential in the treatment of ASD.
Collapse
Affiliation(s)
- Arushi Sandhu
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Kajal Rawat
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Vipasha Gautam
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Antika Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), 4th Floor, Research Block B, Chandigarh 160012, India.
| |
Collapse
|
6
|
Emerging Potential of the Phosphodiesterase (PDE) Inhibitor Ibudilast for Neurodegenerative Diseases: An Update on Preclinical and Clinical Evidence. Molecules 2022; 27:molecules27238448. [PMID: 36500540 PMCID: PMC9737612 DOI: 10.3390/molecules27238448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases constitute a broad range of central nervous system disorders, characterized by neuronal degeneration. Alzheimer's disease, Parkinson's disease, amyolotrophic lateral sclerosis (ALS), and progressive forms of multiple sclerosis (MS) are some of the most frequent neurodegenerative diseases. Despite their diversity, these diseases share some common pathophysiological mechanisms: the abnormal aggregation of disease-related misfolded proteins, autophagosome-lysosome pathway dysregulation, impaired ubiquitin-proteasome system, oxidative damage, mitochondrial dysfunction and excessive neuroinflammation. There is still no effective drug that could halt the progression of neurodegenerative diseases, and the current treatments are mainly symptomatic. In this regard, the development of novel multi-target pharmaceutical approaches presents an attractive therapeutic strategy. Ibudilast, an anti-inflammatory drug firstly developed as an asthma treatment, is a cyclic nucleotide phosphodiesterases (PDEs) inhibitor, which mainly acts by increasing the amount of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), while downregulating the pro-inflammatory factors, such as tumor necrosis factor-α (TNF-α), macrophage migration inhibitory factor (MIF) and Toll-like receptor 4 (TLR-4). The preclinical evidence shows that ibudilast may act neuroprotectively in neurodegenerative diseases, by suppressing neuroinflammation, inhibiting apoptosis, regulating the mitochondrial function and by affecting the ubiquitin-proteasome and autophagosome-lysosome pathways, as well as by attenuating oxidative stress. The clinical trials in ALS and progressive MS also show some promising results. Herein, we aim to provide an update on the emerging preclinical and clinical evidence on the therapeutic potential of ibudilast in these disorders, discuss the potential challenges and suggest the future directions.
Collapse
|
7
|
Philippov I, Gatilov Y, Sonina A, Vorob’ev A. Oxidative [3+2]Cycloaddition of Alkynylphosphonates with Heterocyclic N-Imines: Synthesis of Pyrazolo[1,5- a]Pyridine-3-phosphonates. Molecules 2022; 27:molecules27227913. [PMID: 36432015 PMCID: PMC9694626 DOI: 10.3390/molecules27227913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
A series of pyrazolo[1,5-a]pyridine-3-ylphosphonates were prepared with moderate to good yields by the oxidative [3+2]cycloaddition of 2-subtituted ethynylphosphonates with in situ generated pyridinium-N-imines and their annulated analogs. 2-Aliphatic and 2-Ph acetylenes demonstrate low activity, and the corresponding pyrazolopyridines were achieved with a moderate yield in the presence of 10 mol% Fe(NO3)3·9H2O. At the same time, tetraethyl ethynylbisphosphonate, diethyl 2-TMS- and 2-OPh-ethynylphosphonates possess much greater reactivity and the corresponding pyrazolo[1,5-a]pyridines, and their annulated derivatives were obtained with good to excellent yields without any catalyst. 2-Halogenated ethynylphosphonates also readily reacted with pyridinium-N-imines, forming complex mixtures containing poor amounts of 2-halogenated pyrazolopyridines.
Collapse
Affiliation(s)
- Igor Philippov
- Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| | - Yuriy Gatilov
- Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| | - Alina Sonina
- Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentiev Avenue, 630090 Novosibirsk, Russia
| | - Aleksey Vorob’ev
- Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentiev Avenue, 630090 Novosibirsk, Russia
- Department of Natural Science, Faculty of Organic Chemistry, Novosibirsk State University, 1 Pirogova Street, 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-952-9488714
| |
Collapse
|
8
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
9
|
Hongjin Zhai, Zhang S, Ampomah-Wireko M, Wang H, Cao Y, Yang P, Yang Y, Frejat FOA, Wang L, Zhao B, Ren C, Wu C. Pyrazole: An Important Core in Many Marketed and Clinical Drugs. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022060280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Das S, Bhatia R. Liquid extraction surface analysis-mass spectrometry: An advanced and environment-friendly analytical tool in modern analysis. J Sep Sci 2022; 45:2746-2765. [PMID: 35579471 DOI: 10.1002/jssc.202100996] [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: 12/17/2021] [Revised: 04/23/2022] [Accepted: 05/10/2022] [Indexed: 11/12/2022]
Abstract
The Liquid Extraction Surface Analysis technique is a new high-throughput instrument for ambient mass spectrometry. The benefits of the Liquid Extraction Surface Analysis-Mass Spectrometry approach are the high throughput screening of samples and the absence of sample preparation. Liquid Extraction Surface Analysis-Mass Spectrometry also consumes less solvent for extraction, making it more environmentally friendly and there is no substrate restriction. It utilizes advanced instrumentation like the use of robotic pipettes, nanoelectrospray systems, electronspray ionization chips which makes it highly efficient. In recent years, Liquid Extraction Surface Analysis-Mass Spectrometry has seen widespread use in a variety of analytical fields including drug metabolite analysis, mapping drug distribution in tissues, protein and lipid characterization etc. In this review, we have summarized the basic working principles of the Liquid Extraction Surface Analysis-Mass Spectrometry approach in detail along with a detailed description of the recently reported applications in the analysis of proteins, lipids, drugs and foods. The investigated analytes along with detection methodologies and significant outcomes of various research reports have been presented with the help of tables. This tool has also been utilized in clinical investigations of biological fluids, fingerprint analysis and authentication of agarwood. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shibam Das
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
| |
Collapse
|
11
|
Gharooni AA, Kwon BK, Fehlings MG, Boerger TF, Rodrigues-Pinto R, Koljonen PA, Kurpad SN, Harrop JS, Aarabi B, Rahimi-Movaghar V, Wilson JR, Davies BM, Kotter MRN, Guest JD. Developing Novel Therapies for Degenerative Cervical Myelopathy [AO Spine RECODE-DCM Research Priority Number 7]: Opportunities From Restorative Neurobiology. Global Spine J 2022; 12:109S-121S. [PMID: 35174725 PMCID: PMC8859698 DOI: 10.1177/21925682211052920] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY DESIGN Narrative review. OBJECTIVES To provide an overview of contemporary therapies for the James Lind Alliance priority setting partnership for degenerative cervical myelopathy (DCM) question: 'Can novel therapies, including stem-cell, gene, pharmacological and neuroprotective therapies, be identified to improve the health and wellbeing of people living with DCM and slow down disease progression?' METHODS A review of the literature was conducted to outline the pathophysiology of DCM and present contemporary therapies that may hold therapeutic value in 3 broad categories of neuroprotection, neuroregeneration, and neuromodulation. RESULTS Chronic spinal cord compression leads to ischaemia, neuroinflammation, demyelination, and neuronal loss. Surgical intervention may halt progression and improve symptoms, though the majority do not make a full recovery leading to lifelong disability. Neuroprotective agents disrupt deleterious secondary injury pathways, and one agent, Riluzole, has undergone Phase-III investigation in DCM. Although it did not show efficacy on the primary outcome modified Japanese Orthopaedic Association scale, it showed promising results in pain reduction. Regenerative approaches are in the early stage, with one agent, Ibudilast, currently in a phase-III investigation. Neuromodulation approaches aim to therapeutically alter the state of spinal cord excitation by electrical stimulation with a variety of approaches. Case studies using electrical neuromuscular and spinal cord stimulation have shown positive therapeutic utility. CONCLUSION There is limited research into interventions in the 3 broad areas of neuroprotection, neuroregeneration, and neuromodulation for DCM. Contemporary and novel therapies for DCM are now a top 10 priority, and whilst research in these areas is limited in DCM, it is hoped that this review will encourage research into this priority.
Collapse
Affiliation(s)
- Aref-Ali Gharooni
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - Brian K. Kwon
- Vancouver Spine Surgery Institute, Department of Orthopedics, The University of British Columbia, Vancouver, BC, Canada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Timothy F. Boerger
- Department of Neurosurgery, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Ricardo Rodrigues-Pinto
- Spinal Unit (UVM), Department of Orthopaedics, Centro Hospitalar Universitário do Porto - Hospital de Santo António, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Paul Aarne Koljonen
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - James S. Harrop
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bizhan Aarabi
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vafa Rahimi-Movaghar
- Department of Neurosurgery, Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jefferson R. Wilson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Benjamin M. Davies
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - Mark R. N. Kotter
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - James D. Guest
- Department of Neurosurgery and The Miami Project to Cure Paralysis, The Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
12
|
Liu S, Gao J, Liu K, Zhang HL. Microbiota-gut-brain axis and Alzheimer's disease: Implications of the blood-brain barrier as an intervention target. Mech Ageing Dev 2021; 199:111560. [PMID: 34411603 DOI: 10.1016/j.mad.2021.111560] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/26/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
The microbiota-gut-brain axis has emerged as a focal point of biomedical research. Alterations of gut microbiota are involved in not only various immune/inflammatory disorders but also neurological disorders including Alzheimer's disease (AD). The initial stage of the involvement of gut microbiota in the pathogenesis of AD may be the dysfunction of the blood-brain barrier (BBB). Gut microbiota-derived products in the circulation can worsen the BBB integrity, easily cross the disrupted BBB and enter the brain to promote pathological changes in AD. In this review, we first summarize the current evidence of the associations among gut microbiota, AD, and BBB integrity. We then discuss the mechanism of gut microbiota on BBB dysfunction with a focus on bacteria-derived lipopolysaccharide and exosomal high-mobility group box 1. Novel insights into the modification of the BBB as an intervention approach for AD are highlighted as well.
Collapse
Affiliation(s)
- Shan Liu
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiguo Gao
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Kangding Liu
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China.
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Shuangqing Road 83, 100085, Beijing, China.
| |
Collapse
|
13
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
14
|
Baker D, Hadjicharalambous C, Gnanapavan S, Giovannoni G. Can rheumatologists stop causing demyelinating disease? Mult Scler Relat Disord 2021; 53:103057. [PMID: 34126373 DOI: 10.1016/j.msard.2021.103057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Perhaps the most informative experiments in human disease are clinical trials and notably, responses to specific therapies can be highly-informative to help understand disease pathogenesis. There are reagents that inhibit a variety of different autoimmune conditions, such as CD20 memory B cell depleters that are active in both multiple sclerosis (MS), rheumatoid arthritis (RA) and other conditions, suggesting influences on common immune mechanisms in different diseases. However, a notable exception seemed to be the use of tumour necrosis factor (TNF) inhibitors that limits RA, yet seem to, rarely, trigger demyelination and induce MS. This was first seen with TNF-inhibiting monoclonal antibodies and TNF-receptor-immunoglobulin fusion proteins. However, this is also seen with tyrosine and Janus kinase inhibitors that inhibit RA, yet induce demyelinating disease in some individuals PURPOSE: To provide an overview, from a B cell centric perspective, that may underpin the biology that links arthritis treatments to the development of demyelinating disease. CONCLUSIONS It is apparent that the disease modifying anti-rheumatoid drugs that cause demyelination share a number of common features. These agents tend to inhibit TNF-receptor signalling, augment or exhibit limited inhibitor activity on class-switched memory B cells and importantly appear to be relatively excluded from the central nervous system (CNS). They will thus not target ectopic B cell follicles in the CNS, unlike that occurring in peripheral autoimmunity as seen with anti-TNF treatments in RA. Agents such as ibudilast and some Janus kinase inhibitors that inhibit TNF and clearly penetrate the CNS do not appear to induce demyelination and may even be neuroprotective. It remains to be established whether selection or development of CNS penetrant agents may avoid CNS-complications of treatments for RA. Clearly, further studies are warranted.
Collapse
Affiliation(s)
- David Baker
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, E1 2AT, United Kingdom.
| | - Charalambos Hadjicharalambous
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, E1 2AT, United Kingdom
| | - Sharmilee Gnanapavan
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, E1 2AT, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Gavin Giovannoni
- Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, E1 2AT, United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
15
|
Liu Q, Min T, Dong J, Wang X. Minocycline alleviates the symptoms of morphine withdrawal via the CaMKII-Ras-ERK signaling pathway. Neurosci Lett 2021; 752:135825. [PMID: 33727130 DOI: 10.1016/j.neulet.2021.135825] [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: 11/11/2020] [Revised: 02/18/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate the effect of minocycline on morphine withdrawal symptoms. METHODS We established a rat model of morphine dependence, then injected the animals with naloxone to induce withdrawal symptoms. Minocycline was injected into the midbrain periaqueductal gray and its effect on withdrawal symptoms and Ca2+-dependent protein kinase (CaMKII), Ras, and phospho-extracellular signal-regulated kinase (p-ERK) expression was observed. RESULTS Minocycline inhibited withdrawal symptoms such as "wet dog" shakes, teeth chatter, and ptosis, perhaps by inhibiting the activation of microglia and the expression of CaMKII, Ras, and p-ERK. Minocycline had no effect on the behavior of control rats or on CaMKII, Ras, or p-ERK expression. CONCLUSION Minocycline alleviates morphine withdrawal symptoms by inhibiting the activation of microglia and downregulating the expression of CaMKII, Ras, and p-ERK.
Collapse
Affiliation(s)
- Qiaofeng Liu
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China; Department of Pathogenic Biology, Chengdu Medical College, Chengdu, China
| | - Ting Min
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China; Department of Pathogenic Biology, Chengdu Medical College, Chengdu, China
| | - Jun Dong
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China; Department of Pathogenic Biology, Chengdu Medical College, Chengdu, China
| | - Xin Wang
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China; Department of Pathogenic Biology, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
16
|
Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2021; 1:699-716. [PMID: 22934262 PMCID: PMC3429574 DOI: 10.4161/onci.20696] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are prototypic pattern recognition receptors (PRRs) best known for their ability to activate the innate immune system in response to conserved microbial components such as lipopolysaccharide and double-stranded RNA. Accumulating evidence indicates that the function of TLRs is not restricted to the elicitation of innate immune responses against invading pathogens. TLRs have indeed been shown to participate in tissue repair and injury-induced regeneration as well as in adaptive immune responses against cancer. In particular, TLR4 signaling appears to be required for the efficient processing and cross-presentation of cell-associated tumor antigens by dendritic cells, which de facto underlie optimal therapeutic responses to some anticancer drugs. Thus, TLRs constitute prominent therapeutic targets for the activation/intensification of anticancer immune responses. In line with this notion, long-used preparations such as the Coley toxin (a mixture of killed Streptococcus pyogenes and Serratia marcescens bacteria) and the bacillus Calmette-Guérin (BCG, an attenuated strain of Mycobacterium bovis originally developed as a vaccine against tuberculosis), both of which have been associated with consistent anticancer responses, potently activate TLR2 and TLR4 signaling. Today, besides BCG, only one TLR agonist is FDA-approved for therapeutic use in cancer patients: imiquimod. In this Trial Watch, we will briefly present the role of TLRs in innate and cognate immunity and discuss the progress of clinical studies evaluating the safety and efficacy of experimental TLR agonists as immunostimulatory agents for oncological indications.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Injection of minocycline into the periaqueductal gray attenuates morphine withdrawal signs. Neurosci Lett 2020; 736:135283. [PMID: 32739271 DOI: 10.1016/j.neulet.2020.135283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/07/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
Abstract
This study investigated the effects of minocycline microinjections, into the midbrain periaqueductal gray (PAG), on morphine withdrawal and the expression of pannexin-1 (panx1), phosphorylated mammalian target of rapamycin (p-mTOR), protein kinase A (PKA), and cAMP response element-binding protein (CREB). Rats were injected with morphine, intraperitoneally, at increasing doses, twice per day, to establish animal models of morphine exposure. Minocycline was administered into the PAG before the first intraperitoneal (i.p.) injection of morphine each day, on days 1-4. On the last day of the experiment, all rats were injected with naloxone, and morphine withdrawal was observed, and then changes in the expression levels of ionized calcium-binding adaptor molecule 1 (Iba1) and its downstream factors, panx1, p-mTOR, PKA, and CREB were evaluated by western blot and immunohistochemistry analyses. Morphine withdrawal increased microglial activation, whereas minocycline could inhibit microglial activation and withdrawal and the downregulation of panx1, p-mTOR, PKA, and CREB expression, reducing the effects of morphine withdrawal.
Collapse
|
18
|
Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in numerous functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and inflammation within the central nervous system (neuroinflammation) have been investigated to treat various aspects of drug abuse and dependence. This article presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
19
|
Synthesis of 3-Fluoropyrazolo[1,5-A]Pyridines by Fluorination of Methyl Pyrazolo[1,5-A]Pyridine-3-Carboxylates. Chem Heterocycl Compd (N Y) 2020. [DOI: 10.1007/s10593-020-02757-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Donnelly CR, Andriessen AS, Chen G, Wang K, Jiang C, Maixner W, Ji RR. Central Nervous System Targets: Glial Cell Mechanisms in Chronic Pain. Neurotherapeutics 2020; 17:846-860. [PMID: 32820378 PMCID: PMC7609632 DOI: 10.1007/s13311-020-00905-7] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Interactions between central glial cells and neurons in the pain circuitry are critical contributors to the pathogenesis of chronic pain. In the central nervous system (CNS), two major glial cell types predominate: astrocytes and microglia. Injuries or pathological conditions which evoke pain are concurrently associated with the presence of a reactive microglia or astrocyte state, which is characterized by a variety of changes in the morphological, molecular, and functional properties of these cells. In this review, we highlight the changes that reactive microglia and astrocytes undergo following painful injuries and insults and discuss the critical and interactive role these two cell types play in the initiation and maintenance of chronic pain. Additionally, we focus on several crucial mechanisms by which microglia and astrocytes contribute to chronic pain and provide commentary on the therapeutic promise of targeting these pathways. In particular, we discuss how the inflammasome in activated microglia drives maturation and release of key pro-inflammatory cytokines, which drive pain through neuronal- and glial regulations. Moreover, we highlight several potentially-druggable hemichannels and proteases produced by reactive microglia and astrocytes in pain states and discuss how these pathways regulate distinct phases during pain pathogenesis. We also review two emerging areas in chronic pain research: 1) sexually dimorphic glial cell signaling and 2) the role of oligodendrocytes. Finally, we highlight important considerations for potential pain therapeutics targeting glial cell mediators as well as questions that remain in our conceptual understanding of glial cell activation in pain states.
Collapse
Affiliation(s)
- Christopher R Donnelly
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Amanda S Andriessen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Gang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kaiyuan Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
21
|
Abstract
This chapter describes recent clinical trials for opioid use disorder (OUD), an area that has rapidly accelerated in response to the opioid overdose crisis in the USA and newly appropriated funding. Trials involve a wide range of compounds including cannabinoids and psychedelics, new and existing compounds targeting domains emerging from addiction neuroscience, agents repurposed from other indications, and novel strategies including vaccines, enzymes, and other biologicals. In parallel, new formulations of existing compounds offer immediate promise, as do a variety of web-based interventions and smartphone-delivered apps. Trials focused on implementing existing effective interventions in mainstream healthcare settings, and others focused on special populations, e.g., adolescents, criminal justice, pregnant women, native Americans, etc., have the potential to vastly expand treatment in the near term. Given the range of ongoing and recent trials, this chapter is not intended to be an exhaustive review but rather to present an overview of approaches within the framework of the opioid treatment cascade and the context of current OUD pharmacotherapies.
Collapse
Affiliation(s)
- Esther Blessing
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA.
| | - Sanya Virani
- Department of Psychiatry, Maimonides Medical Center, Brooklyn, NY, USA
| | - John Rotrosen
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
22
|
Baillie GS, Tejeda GS, Kelly MP. Therapeutic targeting of 3',5'-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat Rev Drug Discov 2019; 18:770-796. [PMID: 31388135 PMCID: PMC6773486 DOI: 10.1038/s41573-019-0033-4] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2019] [Indexed: 01/24/2023]
Abstract
Phosphodiesterases (PDEs), enzymes that degrade 3',5'-cyclic nucleotides, are being pursued as therapeutic targets for several diseases, including those affecting the nervous system, the cardiovascular system, fertility, immunity, cancer and metabolism. Clinical development programmes have focused exclusively on catalytic inhibition, which continues to be a strong focus of ongoing drug discovery efforts. However, emerging evidence supports novel strategies to therapeutically target PDE function, including enhancing catalytic activity, normalizing altered compartmentalization and modulating post-translational modifications, as well as the potential use of PDEs as disease biomarkers. Importantly, a more refined appreciation of the intramolecular mechanisms regulating PDE function and trafficking is emerging, making these pioneering drug discovery efforts tractable.
Collapse
Affiliation(s)
- George S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Gonzalo S Tejeda
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, UK
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
23
|
Woodcock EA, Hillmer AT, Mason GF, Cosgrove KP. Imaging Biomarkers of the Neuroimmune System among Substance Use Disorders: A Systematic Review. MOLECULAR NEUROPSYCHIATRY 2019; 5:125-146. [PMID: 31312635 PMCID: PMC6597912 DOI: 10.1159/000499621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
There is tremendous interest in the role of the neuroimmune system and inflammatory processes in substance use disorders (SUDs). Imaging biomarkers of the neuroimmune system in vivo provide a vital translational bridge between preclinical and clinical research. Herein, we examine two imaging techniques that measure putative indices of the neuroimmune system and review their application among SUDs. Positron emission tomography (PET) imaging of 18 kDa translocator protein availability is a marker associated with microglia. Proton magnetic resonance spectroscopy quantification of myo-inositol levels is a putative glial marker found in astrocytes. Neuroinflammatory responses are initiated and maintained by microglia and astrocytes, and thus represent important imaging markers. The goal of this review is to summarize neuroimaging findings from the substance use literature that report data using these markers and discuss possible mechanisms of action. The extant literature indicates abused substances exert diverse and complex neuroimmune effects. Moreover, drug effects may change across addiction stages, i.e. the neuroimmune effects of acute drug administration may differ from chronic use. This burgeoning field has considerable potential to improve our understanding and treatment of SUDs. Future research is needed to determine how targeting the neuroimmune system may improve treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Kelly P. Cosgrove
- Departments of Psychiatry, and of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Zhong S, Zhou Z, Liang Y, Cheng X, Li Y, Teng W, Zhao M, Liu C, Guan M, Zhao C. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol 2019; 45:369-393. [PMID: 31106639 DOI: 10.1080/1040841x.2019.1608905] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a progressive, often irreversible condition that produces severe neurological deficits. Emerging data suggest that chemotherapy also exerts detrimental effects on gut microbiota composition and intestinal permeability, contributing to dysbiosis and inflammation. Compared with other complications associated with chemotherapy, such as diarrhoea and mucositis, CIPN is of particular concern because it is the most common reason for terminating or suspending treatment. However, specific and effective curative treatment strategies are lacking. In this review, we provide an update on current preclinical and clinical understandings about the role of gut microbiota in CIPN. The gut microbiota serves as an intersection between the microbiome-gut-brain and the neuroimmune-endocrine axis, forming a complex network that can directly or indirectly affect key components involved in the manifestations of CIPN. Herein, we discuss several potential mechanisms within the context of the networks and summarize alterations in gut microbiome induced by chemotherapeutic drugs, providing great potential for researchers to target pathways associated with the gut microbiome and overcome CIPN.
Collapse
Affiliation(s)
- Shanshan Zhong
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Zhike Zhou
- Department of Geriatrics, The First Hospital of China Medical University , Shenyang , PR China
| | - Yifan Liang
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Xi Cheng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University , Shenyang , PR China
| | - Weiyu Teng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Mei Zhao
- Department of Cardiology, Shengjing Hospital of China Medical University , Shenyang , PR China
| | - Chang Liu
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Meiting Guan
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Chuansheng Zhao
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| |
Collapse
|
25
|
Ganguly P, Honeycutt JA, Rowe JR, Demaestri C, Brenhouse HC. Effects of early life stress on cocaine conditioning and AMPA receptor composition are sex-specific and driven by TNF. Brain Behav Immun 2019; 78:41-51. [PMID: 30654007 PMCID: PMC6488364 DOI: 10.1016/j.bbi.2019.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/18/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
Exposure to early life adversity can predispose adolescents to the formation of substance abuse disorders. In rodents, early stressors such as repeated maternal separation (MS) impact AMPAR activity in the prefrontal cortex (PFC) and nucleus accumbens (NAc), regions involved in drug-cue association after cocaine-induced conditioned place preference (CPP). Notably, previous reports suggest that the pro-inflammatory cytokine tumor necrosis factor (TNF) regulates AMPAR subunit composition; increased TNF levels are reported to reduce GluA2-positive AMPARs. Since MS can elevate adolescent TNF levels, the stressor may therefore alter AMPAR subunit composition via neuroimmune signaling, thereby affecting cocaine-induced CPP. We tested the specific role of soluble TNF in MS-induced GluA2 loss and cocaine-induced CPP with biologic disruption of TNF signaling. TNF gene and protein expression were elevated in both PFC and NAc of MS males, but not females. GluA2 expression was reduced in both regions in only male MS rats, and systemic treatment with either ibudilast - a phosphodiesterase inhibitor, or XPro1595 - a blood-brain barrier-permeable blocker of soluble TNF - reversed such loss. MS males also formed greater preference for a cocaine-paired environment, the expression of which returned to control levels after XPro1595 administration. These data suggest a sex-specific mechanistic link between TNF signaling and changes in GluA2 expression and drug-cue conditioning, thereby providing further evidence for a role of MS and neuro-immune activity in cortical and striatal AMPAR changes. Moreover, manipulation of the TNF signaling pathway represents a novel approach for influencing response to reinforcing effects of drug use.
Collapse
Affiliation(s)
- Prabarna Ganguly
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Jennifer A Honeycutt
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - June R Rowe
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Camila Demaestri
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Heather C Brenhouse
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Clanchy FIL, Williams RO. Ibudilast Inhibits Chemokine Expression in Rheumatoid Arthritis Synovial Fibroblasts and Exhibits Immunomodulatory Activity in Experimental Arthritis. Arthritis Rheumatol 2019; 71:703-711. [PMID: 30474934 DOI: 10.1002/art.40787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/20/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Ibudilast is a well-tolerated, orally available phosphodiesterase 4 (PDE4) inhibitor used to treat asthma and stroke. Since PDE4 inhibition suppresses inflammatory mediator production and cell proliferation in leukocytes, ibudilast may be a valuable therapy for the treatment of inflammatory autoimmune diseases such as rheumatoid arthritis (RA). This study was undertaken to assess the therapeutic potential of ibudilast by measuring its capacity to modulate inflammation in human leukocytes and RA synovial fibroblasts (RASFs) and in experimental arthritis. METHODS Using standard curve quantitative polymerase chain reaction, the effect of ibudilast on gene expression in activated human leukocytes and RASFs was measured. Ibudilast was used to treat DBA/1 mice with collagen-induced arthritis, and an adoptive transfer model was used to assess its tolerogenic capacity. RESULTS Ibudilast inhibited the expression of TNF, IL12A, and IL12B and the secretion of tumor necrosis factor (TNF) and interleukin-12 (IL-12)/23p40 from leukocytes, and reduced the expression of CCL5 and CCL3 in activated RASFs. Treatment of experimental arthritis with ibudilast resulted in a reduction in IL-17-producing cells and inhibition of disease progression. When combined with a TNF inhibitor, ibudilast caused marked suppression of active disease. Exposure of leukocytes from type II collagen-immunized DBA/1 mice to ibudilast in vitro attenuated their ability to adoptively transfer arthritis to DBA/1J-PrkdcSCID mice, providing evidence of an immunomodulatory effect. CONCLUSION Our findings indicate that ibudilast reduces the expression and/or secretion of inflammatory mediators from activated human leukocytes and RASFs, inhibits Th17 cell responses in vivo, and improves established arthritis. Given the established safety profile of ibudilast in humans, its clinical evaluation in RA, either alone or in combination with a TNF inhibitor, should be considered.
Collapse
|
27
|
Human Toll-Like Receptor 4 (hTLR4): Structural and functional dynamics in cancer. Int J Biol Macromol 2019; 122:425-451. [DOI: 10.1016/j.ijbiomac.2018.10.142] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/10/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
|
28
|
Bruno K, Woller SA, Miller YI, Yaksh TL, Wallace M, Beaton G, Chakravarthy K. Targeting toll-like receptor-4 (TLR4)-an emerging therapeutic target for persistent pain states. Pain 2018; 159:1908-1915. [PMID: 29889119 PMCID: PMC7890571 DOI: 10.1097/j.pain.0000000000001306] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors that initiate signaling in innate and adaptive immune pathways. The highly conserved family of transmembrane proteins comprises an extracellular domain that recognizes exogenous and endogenous danger molecules and an ectodomain that activates downstream pathways in response. Recent studies suggest that continuous activation or dysregulation of TLR signaling may contribute to chronic disease states. The receptor is located not only on inflammatory cells (meningeal and peripheral macrophages) but on neuraxial glia (microglia and astrocytes), Schwann cells, fibroblasts, dorsal root ganglia, and dorsal horn neurons. Procedures blocking TLR functionality have shown pronounced effects on pain behavior otherwise observed in models of chronic inflammation and nerve injury. This review addresses the role of TLR4 as an emerging therapeutic target for the evolution of persistent pain and its role in noncanonical signaling, mediating anomalous pro-algesic actions of opiates. Accordingly, molecules targeting inhibition of this receptor have promise as disease-modifying and opioid-sparing alternatives for persistent pain states.
Collapse
Affiliation(s)
- Kelly Bruno
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Center for Excellence in Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | - Sarah A. Woller
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
| | - Yury I. Miller
- Department of Medicine, University of California San Diego Health Science, La Jolla, CA, USA
| | - Tony L. Yaksh
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Mark Wallace
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Graham Beaton
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| | - Krishnan Chakravarthy
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Douleur Therapeutics, 10225 Barnes Canyon Road, Suite A104, San Diego, CA, USA
| |
Collapse
|
29
|
Litten RZ, Falk DE, Ryan ML, Fertig J, Leggio L. Advances in Pharmacotherapy Development: Human Clinical Studies. Handb Exp Pharmacol 2018; 248:579-613. [PMID: 29294197 DOI: 10.1007/164_2017_79] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
For more than 25 years, researchers have made advances in developing medications to treat alcohol use disorder (AUD), highlighted by the US Food and Drug Administration's (FDA's) approval of disulfiram, naltrexone (oral and long-acting), and acamprosate. These medications are also approved in Europe, where the European Medicines Agency (EMA) recently added a fourth medication, nalmefene, for AUD. Despite these advances, today's medications have a small effect size, showing efficacy for only a limited number of individuals with AUD. However, a host of new medications, which act on variety of pharmacologic targets, are in the pipeline and have been evaluated in numerous human studies. This article reviews the efficacy and safety of medications currently being tested in human trials and looks at ongoing efforts to identify candidate compounds in human studies. As mentioned in the National Institute on Alcohol Abuse and Alcoholism's Strategic Plan 2017-2021 ( https://www.niaaa.nih.gov/sites/default/files/StrategicPlan_NIAAA_optimized_2017-2020.pdf ), medications development remains a high priority. By developing more effective and safe medications, and identifying those patients who will benefit the most from these treatments, we can provide clinicians with the tools they need to treat this devastating disorder, providing relief for patients and their families and markedly improving public health and safety.
Collapse
Affiliation(s)
- Raye Z Litten
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| | - Daniel E Falk
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Megan L Ryan
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Joanne Fertig
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Lorenzo Leggio
- Section of Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, Bethesda, MD, USA
| |
Collapse
|
30
|
Affiliation(s)
- Nicole E Burma
- Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, Physiology & Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Charlie Ht Kwok
- Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, Physiology & Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tuan Trang
- Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, Physiology & Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
31
|
Ibudilast reduces oxaliplatin-induced tactile allodynia and cognitive impairments in rats. Behav Brain Res 2017; 334:109-118. [DOI: 10.1016/j.bbr.2017.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 01/05/2023]
|
32
|
Ray LA, Bujarski S, Shoptaw S, Roche DJO, Heinzerling K, Miotto K. Development of the Neuroimmune Modulator Ibudilast for the Treatment of Alcoholism: A Randomized, Placebo-Controlled, Human Laboratory Trial. Neuropsychopharmacology 2017; 42:1776-1788. [PMID: 28091532 PMCID: PMC5520778 DOI: 10.1038/npp.2017.10] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
Abstract
Current directions in medication development for alcohol use disorder (AUD) emphasize the need to identify novel molecular targets and efficiently screen new compounds aimed at those targets. Ibudilast (IBUD) is a neuroimmune modulator that inhibits phosphodiesterase-4 and -10 and macrophage migration inhibitory factor and was recently found to reduce alcohol intake in rats by ∼50%. To advance medication development for AUD, the present study consists of a randomized, crossover, double-blind, placebo-controlled laboratory study of IBUD in nontreatment-seeking individuals with current (ie, past month) mild-to-severe AUD. This study tested the safety, tolerability, and initial human laboratory efficacy of IBUD (50 mg b.i.d.) on primary measures of subjective response to alcohol as well as secondary measures of cue- and stress-induced changes in craving and mood. Participants (N=24) completed two separate 7-day intensive outpatient protocols that included daily visits for medication administration and testing. Upon reaching a stable target dose of IBUD (or matched placebo), participants completed a stress-exposure session (day 5; PM), an alcohol cue-exposure session (day 6; AM), and an i.v. alcohol administration session (day 6; PM). Participants stayed overnight after the alcohol administration, and discharge occurred on day 7 of the protocol. Medication conditions were separated by a washout period that was ⩾7 days. IBUD was well tolerated; however, there were no medication effects on primary measures of subjective response to alcohol. IBUD was associated with mood improvements on the secondary measures of stress exposure and alcohol cue exposure, as well as reductions in tonic levels of craving. Exploratory analyses revealed that among individuals with higher depressive symptomatology, IBUD attenuated the stimulant and mood-altering effects of alcohol as compared with placebo. Together, these findings extend preclinical demonstrations of the potential utility of IBUD for the treatment of AUD and suggest that depressive symptomatology should be considered as a potential moderator of efficacy for pharmacotherapies with neuroimmune effects, such as IBUD.
Collapse
Affiliation(s)
- Lara A Ray
- Department of Psychology, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Steve Shoptaw
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Department of Family Medicine, University of California, Los Angeles, CA, USA
| | - Daniel JO Roche
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Keith Heinzerling
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
- Department of Family Medicine, University of California, Los Angeles, CA, USA
| | - Karen Miotto
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
33
|
Metz VE, Jones JD, Manubay J, Sullivan MA, Mogali S, Segoshi A, Madera G, Johnson KW, Comer SD. Effects of Ibudilast on the Subjective, Reinforcing, and Analgesic Effects of Oxycodone in Recently Detoxified Adults with Opioid Dependence. Neuropsychopharmacology 2017; 42:1825-1832. [PMID: 28393896 PMCID: PMC5520790 DOI: 10.1038/npp.2017.70] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 04/01/2017] [Accepted: 04/03/2017] [Indexed: 11/09/2022]
Abstract
Ibudilast, a nonselective phosphodiesterase inhibitor, is used clinically in Asia for the treatment of asthma and poststroke dizziness. Recent preclinical studies have suggested that it also inhibits glial cell activation in rodents, and may alter opioid-mediated effects, including analgesia and withdrawal symptoms. The effects of ibudilast on the abuse potential of opioids in humans are largely unknown. The present study was designed to examine the influence of ibudilast on subjective (including drug craving), reinforcing, and analgesic effects of oxycodone in human volunteers diagnosed with opioid dependence (equivalent to moderate-severe opioid use disorder). Non-treatment-seeking opioid-dependent male volunteers (n=11) underwent an in-patient detoxification with morphine, followed by maintenance on placebo (0 mg b.i.d.) and active ibudilast (50 mg b.i.d.). Under each maintenance dose, six experimental sample and choice sessions were completed involving oral oxycodone administration (0, 15, and 30 mg/70 kg, p.o.). Subjective effects of oxycodone and drug craving were measured with visual analog scales (VAS) and a Drug Effects Questionnaire. The cold pressor test was used to produce pain, and a modified progressive-ratio choice procedure was used to measure the reinforcing effects of oxycodone. Under the active ibudilast condition compared with the placebo condition, ratings of drug liking following 15 mg of oxycodone were decreased significantly. The mean drug breakpoint value was also significantly lower in the active vs the placebo ibudilast condition under the 15 mg oxycodone condition, but not significantly lower under the 30 mg oxycodone condition. Heroin craving was significantly reduced under active ibudilast vs placebo, and similar effects were observed for tobacco and cocaine craving. Furthermore, mean subjective ratings of pain were lower in the active ibudilast condition. Our data suggest that ibudilast may be useful for treating opioid use disorders and it may enhance the analgesic effects of oxycodone.
Collapse
Affiliation(s)
- Verena E Metz
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA,Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA, Tel: +1 646 774 6139; Fax: +1 646 774 6018; E-mail: or
| | - Jermaine D Jones
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Jeanne Manubay
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Maria A Sullivan
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA,Clinical Research and Development, Alkermes, Waltham, MA, USA
| | - Shanthi Mogali
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Andrew Segoshi
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Gabriela Madera
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | | | - Sandra D Comer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
34
|
Schwenkgrub J, Zaremba M, Joniec-Maciejak I, Cudna A, Mirowska-Guzel D, Kurkowska-Jastrzębska I. The phosphodiesterase inhibitor, ibudilast, attenuates neuroinflammation in the MPTP model of Parkinson's disease. PLoS One 2017; 12:e0182019. [PMID: 28753652 PMCID: PMC5533435 DOI: 10.1371/journal.pone.0182019] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 07/11/2017] [Indexed: 12/22/2022] Open
Abstract
Background/Aims Since the degeneration of the nigrostriatal dopaminergic pathway in Parkinson’s disease (PD) is associated with the inflammation process and decreased levels of cyclic nucleotides, inhibition of up-regulated cyclic nucleotide phosphodiesterases (PDEs) appears to be a promising therapeutic strategy. We used ibudilast (IBD), a non-selective PDE3,4,10,11 inhibitor, due to the abundant PDE 4 and 10 expression in the striatum. The present study for the first time examined the efficacy of IBD in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Methods IBD [0, 20, 30, 40, or 50 mg/kg] was injected b.i.d. subcutaneously for nine days to three-month-old male C57Bl/10Tar mice, beginning two days prior to MPTP (60 mg/kg) intoxication. High-pressure liquid chromatography, Western blot analysis, and real time RT-PCR methods were applied. Results Our study demonstrated that chronic administration of IBD attenuated astroglial reactivity and increased glial cell-derived neurotrophic factor (GDNF) production in the striatum. Moreover, IBD reduced TNF-α, IL-6, and IL-1β expression. Conclusion IBD had a well-defined effect on astroglial activation in the mouse model of PD; however, there was no protective effect in the acute phase of injury. Diminished inflammation and an increased level of GDNF may provide a better outcome in the later stages of neurodegeneration.
Collapse
Affiliation(s)
- Joanna Schwenkgrub
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Zaremba
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
- Laboratory of Magnetic Resonance Imaging of Small Animals, Mossakowski Medical Research Centre, PAS, Warsaw, Poland
- * E-mail:
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudna
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
- 2 Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
35
|
Burma NE, Leduc-Pessah H, Trang T. Genetic deletion of microglial Panx1 attenuates morphine withdrawal, but not analgesic tolerance or hyperalgesia in mice. Channels (Austin) 2017; 11:487-494. [PMID: 28745932 DOI: 10.1080/19336950.2017.1359361] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Opioids are among the most powerful analgesics for managing pain, yet their repeated use can lead to the development of severe adverse effects. In a recent study, we identified the microglial pannexin-1 channel (Panx1) as a critical substrate for opioid withdrawal. Here, we investigated whether microglial Panx1 contributes to opioid-induced hyperalgesia (OIH) and opioid analgesic tolerance using mice with a tamoxifen-inducible deletion of microglial Panx1. We determined that escalating doses of morphine resulted in thermal pain hypersensitivity in both Panx1-expressing and microglial Panx1-deficient mice. In microglial Panx1-deficient mice, we also found that acute morphine antinociception remained intact, and repeated morphine treatment at a constant dose resulted in a progressive decline in morphine antinociception and a reduction in morphine potency. This reduction in morphine antinociceptive potency was indistinguishable from that observed in Panx1-expressing mice. Notably, morphine tolerant animals displayed increased spinal microglial reactivity, but no change of microglial Panx1 expression. Collectively, our findings indicate microglial Panx1 differentially contributes to opioid withdrawal, but not the development of opioid-induced hyperalgesia or tolerance.
Collapse
Affiliation(s)
- Nicole E Burma
- a Comparative Biology and Experimental Medicine , University of Calgary , Calgary, Alberta , Canada.,b Physiology and Pharmacology, Hotchkiss Brain Institute , University of Calgary , Calgary, Alberta , Canada
| | - Heather Leduc-Pessah
- a Comparative Biology and Experimental Medicine , University of Calgary , Calgary, Alberta , Canada.,b Physiology and Pharmacology, Hotchkiss Brain Institute , University of Calgary , Calgary, Alberta , Canada
| | - Tuan Trang
- a Comparative Biology and Experimental Medicine , University of Calgary , Calgary, Alberta , Canada.,b Physiology and Pharmacology, Hotchkiss Brain Institute , University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
36
|
Zerdazi EH, Oliveira J, Vorspan F, Bennabi M, Jamain S, Etain B, Leboyer M, Tamouza R, Bellivier F. TLR4 gene polymorphism associated with lifetime cigarette smoking in bipolar disorder. J Neuroimmunol 2017; 305:96-101. [DOI: 10.1016/j.jneuroim.2017.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
|
37
|
Wieseler J, Ellis A, McFadden A, Stone K, Brown K, Cady S, Bastos LF, Sprunger D, Rezvani N, Johnson K, Rice KC, Maier SF, Watkins LR. Supradural inflammatory soup in awake and freely moving rats induces facial allodynia that is blocked by putative immune modulators. Brain Res 2017; 1664:87-94. [PMID: 28322750 DOI: 10.1016/j.brainres.2017.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 12/25/2022]
Abstract
Facial allodynia is a migraine symptom that is generally considered to represent a pivotal point in migraine progression. Treatment before development of facial allodynia tends to be more successful than treatment afterwards. As such, understanding the underlying mechanisms of facial allodynia may lead to a better understanding of the mechanisms underlying migraine. Migraine facial allodynia is modeled by applying inflammatory soup (histamine, bradykinin, serotonin, prostaglandin E2) over the dura. Whether glial and/or immune activation contributes to such pain is unknown. Here we tested if trigeminal nucleus caudalis (Sp5C) glial and/or immune cells are activated following supradural inflammatory soup, and if putative glial/immune inhibitors suppress the consequent facial allodynia. Inflammatory soup was administered via bilateral indwelling supradural catheters in freely moving rats, inducing robust and reliable facial allodynia. Gene expression for microglial/macrophage activation markers, interleukin-1β, and tumor necrosis factor-α increased following inflammatory soup along with robust expression of facial allodynia. This provided the basis for pursuing studies of the behavioral effects of 3 diverse immunomodulatory drugs on facial allodynia. Pretreatment with either of two compounds broadly used as putative glial/immune inhibitors (minocycline, ibudilast) prevented the development of facial allodynia, as did treatment after supradural inflammatory soup but prior to the expression of facial allodynia. Lastly, the toll-like receptor 4 (TLR4) antagonist (+)-naltrexone likewise blocked development of facial allodynia after supradural inflammatory soup. Taken together, these exploratory data support that activated glia and/or immune cells may drive the development of facial allodynia in response to supradural inflammatory soup in unanesthetized male rats.
Collapse
Affiliation(s)
- Julie Wieseler
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Amanda Ellis
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Andrew McFadden
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Kendra Stone
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Kimberley Brown
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Sara Cady
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Leandro F Bastos
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, CEP 31270-901 Minas Gerais, Brazil
| | - David Sprunger
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Niloofar Rezvani
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Kirk Johnson
- MediciNova Inc, 4350 La Jolla Village Dr., #950, San Diego, CA, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| |
Collapse
|
38
|
IL-4 mediated by HSV vector suppresses morphine withdrawal response and decreases TNFα, NR2B, and pC/EBPβ in the periaqueductal gray in rats. Gene Ther 2017; 24:224-233. [DOI: 10.1038/gt.2017.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/19/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023]
|
39
|
Montesinos J, Alfonso-Loeches S, Guerri C. Impact of the Innate Immune Response in the Actions of Ethanol on the Central Nervous System. Alcohol Clin Exp Res 2016; 40:2260-2270. [PMID: 27650785 DOI: 10.1111/acer.13208] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022]
Abstract
The innate immune response in the central nervous system (CNS) participates in both synaptic plasticity and neural damage. Emerging evidence from human and animal studies supports the role of the neuroimmune system response in many actions of ethanol (EtOH) on the CNS. Research studies have shown that alcohol stimulates brain immune cells, microglia, and astrocytes, by activating innate immune receptors Toll-like receptors (TLRs) and NOD-like receptors (inflammasome NLRs) triggering signaling pathways, which culminate in the production of pro-inflammatory cytokines and chemokines that lead to neuroinflammation. This review focuses on evidence that indicates the participation of TLRs and the inflammasome NLRs signaling response in many effects of EtOH on the CNS, such as neuroinflammation associated with brain damage, cognitive and behavioral dysfunction, and adolescent brain development alterations. It also reviews findings that indicate the role of TLR4-dependent signaling immune molecules in alcohol consumption, reward, and addiction. The research data suggest that overactivation of TLR4 or NLRs increases pro-inflammatory cytokines and mediators to cause neural damage in the cerebral cortex and hippocampus, while modest TLR4 activation, along with the generation of certain cytokines and chemokines in specific brain areas (e.g., amygdala, ventral tegmental area), modulate neurotransmission, alcohol drinking, and alcohol rewards. Elimination of TLR4 and NLRP3 abolishes many neuroimmune effects of EtOH. Despite much progress being made in this area, there are some research gaps and unanswered questions that this review discusses. Finally, potential therapies that target neuroimmune pathways to treat neuropathological and behavioral consequences of alcohol abuse are also evaluated.
Collapse
Affiliation(s)
- Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Silvia Alfonso-Loeches
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain.
| |
Collapse
|
40
|
Ruiz-Pérez D, Benito J, Polo G, Largo C, Aguado D, Sanz L, Gómez de Segura IA. The Effects of the Toll-Like Receptor 4 Antagonist, Ibudilast, on Sevoflurane's Minimum Alveolar Concentration and the Delayed Remifentanil-Induced Increase in the Minimum Alveolar Concentration in Rats. Anesth Analg 2016; 122:1370-6. [PMID: 26859874 DOI: 10.1213/ane.0000000000001171] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Ultralow doses of naloxone, an opioid and toll-like receptor 4 antagonist, blocked remifentanil-induced hyperalgesia and the associated increase in the minimum alveolar concentration (MAC), but not tolerance. The aim was to determine the effects of the toll-like receptor 4 antagonist, ibudilast, on the MAC in the rat and how it might prevent the effects of remifentanil. METHODS Male Wistar rats were randomly allocated to 5 treatment groups (n = 7 per group): 10 mg/kg ibudilast intraperitoneally, 240 µg/kg/h remifentanil IV, ibudilast plus remifentanil, remifentanil plus naloxone IV, or saline. The sevoflurane MAC was determined 3 times in every rat and every day (days 0, 2, and 4): baseline (MAC-A) and 2 further determinations were made after treatments, 1.5 hours apart (MAC-B and MAC-C). RESULTS A reduction in baseline MAC was produced on day 0 by ibudilast, remifentanil, remifentanil plus ibudilast, remifentanil plus naloxone (P < 0.01), but not saline. Similar effects were found on days 2 and 4. A tolerance to remifentanil was found on days 0, 2, and 4, which neither ibudilast nor naloxone prevented. The MAC increase produced by remifentanil on day 4 (P = 0.001) was prevented by either ibudilast or naloxone. CONCLUSIONS Ibudilast, besides reducing the MAC, prevented the delayed increase in baseline MAC produced by remifentanil but not the increase in MAC caused by tolerance to remifentanil.
Collapse
Affiliation(s)
- Daniel Ruiz-Pérez
- From the *Comparative Pain Research Group, Department of Animal Medicine and Surgery, Veterinary Faculty, Complutense University of Madrid (UCM), Madrid, Spain; †Clinical Service of Anesthesia, Faculty of Veterinary Medicine, Department of Clinical Sciences, University of Montreal (UdM), Saint-Hyacinthe, Quebec, Canada; ‡Experimental Surgery Unit, La Paz University Hospital (HULP), Madrid, Spain; and §Mathematics Faculty, Department of Statistics and Operations Research, Complutense University of Madrid (UCM), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
41
|
Fox RJ, Coffey CS, Cudkowicz ME, Gleason T, Goodman A, Klawiter EC, Matsuda K, McGovern M, Conwit R, Naismith R, Ashokkumar A, Bermel R, Ecklund D, Koepp M, Long J, Natarajan S, Ramachandran S, Skaramagas T, Thornell B, Yankey J, Agius M, Bashir K, Cohen B, Coyle P, Delgado S, Dewitt D, Flores A, Giesser B, Goldman M, Jubelt B, Lava N, Lynch S, Miravalle A, Moses H, Ontaneda D, Perumal J, Racke M, Repovic P, Riley C, Severson C, Shinnar S, Suski V, Weinstock-Gutman B, Yadav V, Zabeti A. Design, rationale, and baseline characteristics of the randomized double-blind phase II clinical trial of ibudilast in progressive multiple sclerosis. Contemp Clin Trials 2016; 50:166-77. [PMID: 27521810 DOI: 10.1016/j.cct.2016.08.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/02/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Primary and secondary progressive multiple sclerosis (MS), collectively called progressive multiple sclerosis (PMS), is characterized by gradual progression of disability. The current anti-inflammatory treatments for MS have little or no efficacy in PMS in the absence of obvious active inflammation. Optimal biomarkers for phase II PMS trials is unknown. Ibudilast is an inhibitor of macrophage migration inhibitor factor and phosphodiesterases-4 and -10 and exhibits possible neuroprotective properties. The goals of SPRINT-MS study are to evaluate the safety and efficacy of ibudilast in PMS and to directly compare several imaging metrics for utility in PMS trials. METHODS SPRINT-MS is a randomized, placebo-controlled, phase II trial of ibudilast in patients with PMS. Eligible subjects were randomized 1:1 to receive either ibudilast (100mg/day) or placebo for 96weeks. Imaging is conducted every 24weeks for whole brain atrophy, magnetization transfer ratio, diffusion tensor imaging, cortical brain atrophy, and retinal nerve fiber layer thickness. Clinical outcomes include neurologic disability and patient reported quality of life. Safety assessments include laboratory testing, electrocardiography, and suicidality screening. RESULTS A total of 331 subjects were enrolled, of which 255 were randomized onto active study treatment. Randomized subjects were 53.7% female and mean age 55.7 (SD 7.3) years. The last subject is projected to complete the study in May 2017. CONCLUSION SPRINT-MS is designed to evaluate the safety and efficacy of ibudilast as a treatment for PMS while simultaneously validating five different imaging biomarkers as outcome metrics for use in future phase II proof-of-concept PMS trials.
Collapse
Affiliation(s)
- Robert J Fox
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States.
| | - Christopher S Coffey
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Merit E Cudkowicz
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | | | - Andrew Goodman
- University of Rochester Medical Center, Rochester, NY, United States
| | | | | | - Michelle McGovern
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | - Robin Conwit
- National Institutes of Neurological Disease and Stroke, Bethesda, MD, United States
| | - Robert Naismith
- Washington University School of Medicine, St. Louis, MO, United States
| | - Akshata Ashokkumar
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Robert Bermel
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Dixie Ecklund
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Maxine Koepp
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Jeffrey Long
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Sneha Natarajan
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | | | - Thomai Skaramagas
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Brenda Thornell
- Clinical Coordinating Center, NeuroNEXT, Harvard Partners, Boston, MA, United States
| | - Jon Yankey
- Data Coordinating Center, NeuroNEXT, University of Iowa, Iowa City, IA, United States
| | - Mark Agius
- University of California at Davis, Sacramento, CA; currently at Barrows Neurological Institute, Phoenix, AZ, United States
| | - Khurram Bashir
- University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bruce Cohen
- Northwestern University, Chicago, IL, United States
| | - Patricia Coyle
- State University of New York, Stony Brook, NY, United States
| | - Silvia Delgado
- University of Miami School of Medicine, Miami, FL, United States
| | - Dana Dewitt
- University of Utah, Salt Lake City, UT, United States
| | - Angela Flores
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Barbara Giesser
- University of California at Los Angeles, Los Angeles, CA, United States
| | - Myla Goldman
- University of Virginia at Charlottesville, Charlottesville, VA, United States
| | - Burk Jubelt
- State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Neil Lava
- Emory University, Atlanta, GA, United States
| | - Sharon Lynch
- University of Kansas Medical Center, Kansas City, KS, United States
| | | | - Harold Moses
- Vanderbilt University, Nashville, TN, United States
| | - Daniel Ontaneda
- Cleveland Clinic, Neurological Institute, Cleveland, OH, United States
| | - Jai Perumal
- Weill Cornell Medical College, New York, NY, United States
| | - Michael Racke
- The Ohio State University, Columbus, OH, United States
| | - Pavle Repovic
- Swedish Medical Center at Seattle, Seattle, WA, United States
| | - Claire Riley
- Columbia University Medical Center, New York, NY, United States
| | | | | | - Valerie Suski
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | | | - Vijayshree Yadav
- Oregon Health and Science University, Portland, OR, United States
| | - Aram Zabeti
- University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
42
|
Anwar MA, Al Shehabi TS, Eid AH. Inflammogenesis of Secondary Spinal Cord Injury. Front Cell Neurosci 2016; 10:98. [PMID: 27147970 PMCID: PMC4829593 DOI: 10.3389/fncel.2016.00098] [Citation(s) in RCA: 298] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/30/2016] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) and spinal infarction lead to neurological complications and eventually to paraplegia or quadriplegia. These extremely debilitating conditions are major contributors to morbidity. Our understanding of SCI has certainly increased during the last decade, but remains far from clear. SCI consists of two defined phases: the initial impact causes primary injury, which is followed by a prolonged secondary injury consisting of evolving sub-phases that may last for years. The underlying pathophysiological mechanisms driving this condition are complex. Derangement of the vasculature is a notable feature of the pathology of SCI. In particular, an important component of SCI is the ischemia-reperfusion injury (IRI) that leads to endothelial dysfunction and changes in vascular permeability. Indeed, together with endothelial cell damage and failure in homeostasis, ischemia reperfusion injury triggers full-blown inflammatory cascades arising from activation of residential innate immune cells (microglia and astrocytes) and infiltrating leukocytes (neutrophils and macrophages). These inflammatory cells release neurotoxins (proinflammatory cytokines and chemokines, free radicals, excitotoxic amino acids, nitric oxide (NO)), all of which partake in axonal and neuronal deficit. Therefore, our review considers the recent advances in SCI mechanisms, whereby it becomes clear that SCI is a heterogeneous condition. Hence, this leads towards evidence of a restorative approach based on monotherapy with multiple targets or combinatorial treatment. Moreover, from evaluation of the existing literature, it appears that there is an urgent requirement for multi-centered, randomized trials for a large patient population. These clinical studies would offer an opportunity in stratifying SCI patients at high risk and selecting appropriate, optimal therapeutic regimens for personalized medicine.
Collapse
Affiliation(s)
- M Akhtar Anwar
- Department of Biological and Environmental Sciences, Qatar University Doha, Qatar
| | | | - Ali H Eid
- Department of Biological and Environmental Sciences, Qatar UniversityDoha, Qatar; Department of Pharmacology and Toxicology, Faculty of Medicine, American University of BeirutBeirut, Lebanon
| |
Collapse
|
43
|
Swales JG, Tucker JW, Spreadborough MJ, Iverson SL, Clench MR, Webborn PJH, Goodwin RJA. Mapping drug distribution in brain tissue using liquid extraction surface analysis mass spectrometry imaging. Anal Chem 2015; 87:10146-52. [PMID: 26350423 DOI: 10.1021/acs.analchem.5b02998] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Liquid extraction surface analysis mass spectrometry (LESA-MS) is a surface sampling technique that incorporates liquid extraction from the surface of tissue sections with nanoelectrospray mass spectrometry. Traditional tissue analysis techniques usually require homogenization of the sample prior to analysis via high-performance liquid chromatography mass spectrometry (HPLC-MS), but an intrinsic weakness of this is a loss of all spatial information and the inability of the technique to distinguish between actual tissue penetration and response caused by residual blood contamination. LESA-MS, in contrast, has the ability to spatially resolve drug distributions and has historically been used to profile discrete spots on the surface of tissue sections. Here, we use the technique as a mass spectrometry imaging (MSI) tool, extracting points at 1 mm spatial resolution across tissue sections to build an image of xenobiotic and endogenous compound distribution to assess drug blood-brain barrier penetration into brain tissue. A selection of penetrant and "nonpenetrant" drugs were dosed to rats via oral and intravenous administration. Whole brains were snap-frozen at necropsy and were subsequently sectioned prior to analysis by matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) and LESA-MSI. MALDI-MSI, as expected, was shown to effectively map the distribution of brain penetrative compounds but lacked sufficient sensitivity when compounds were marginally penetrative. LESA-MSI was used to effectively map the distribution of these poorly penetrative compounds, highlighting its value as a complementary technique to MALDI-MSI. The technique also showed benefits when compared to traditional homogenization, particularly for drugs that were considered nonpenetrant by homogenization but were shown to have a measurable penetration using LESA-MSI.
Collapse
Affiliation(s)
- John G Swales
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K.,Biomedical Research Centre, Sheffield Hallam University , Howard Street, Sheffield, South Yorkshire S1 1WB, U.K
| | - James W Tucker
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K
| | - Michael J Spreadborough
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K
| | - Suzanne L Iverson
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K
| | - Malcolm R Clench
- Biomedical Research Centre, Sheffield Hallam University , Howard Street, Sheffield, South Yorkshire S1 1WB, U.K
| | - Peter J H Webborn
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K
| | - Richard J A Goodwin
- Drug Safety and Metabolism, AstraZeneca R&D , Cambridge Science Park, Milton Road, Cambridge CB4 0WG, U.K
| |
Collapse
|
44
|
Johnson JL, Kwok YH, Sumracki NM, Swift JE, Hutchinson MR, Johnson K, Williams DB, Tuke J, Rolan PE. Glial Attenuation With Ibudilast in the Treatment of Medication Overuse Headache: A Double-Blind, Randomized, Placebo-Controlled Pilot Trial of Efficacy and Safety. Headache 2015; 55:1192-208. [DOI: 10.1111/head.12655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2015] [Indexed: 01/08/2023]
Affiliation(s)
| | - Yuen H. Kwok
- Discipline of Pharmacology, University of Adelaide; Adelaide Australia
| | | | - James E. Swift
- Discipline of Pharmacology, University of Adelaide; Adelaide Australia
| | | | | | - Desmond B. Williams
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide Australia
| | - Jonathon Tuke
- School of Mathematical Sciences; University of Adelaide; Adelaide Australia
| | - Paul E. Rolan
- Discipline of Pharmacology, University of Adelaide; Adelaide Australia
- Pain and Anaesthesia Research Clinic, Royal Adelaide Hospital; Adelaide Australia
- Pain Management Unit, Royal Adelaide Hospital; Adelaide Australia
| |
Collapse
|
45
|
Ibudilast reverses the decrease in the synaptic signaling protein phosphatidylethanolamine-binding protein 1 (PEBP1) produced by chronic methamphetamine intake in rats. Drug Alcohol Depend 2015; 152:15-23. [PMID: 25962787 DOI: 10.1016/j.drugalcdep.2015.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND Chronic methamphetamine intake has been shown to induce a neuroinflammatory state leading to significant changes in brain functioning including behavioral changes. These changes can persist for years after drug use is discontinued and likely contribute to the risk of relapse. A better understanding of inflammation responses associated with methamphetamine intake may help in designing novel and more efficacious treatment strategies. METHODS Rats were trained to self-administer methamphetamine or saline on a variable ratio 3 schedule of reinforcement (25 days). This training was followed by 12 days of extinction (i.e., methamphetamine unavailable) during which rats received daily post-session administration of ibudilast (AV411; 2.5 or 7.5mg/kg) or saline. Following extinction, synaptosomes were isolated from the prefrontal cortex (PFC) and the differential pattern of synaptic proteins was assessed using mass spectrometry based proteomics. RESULTS Treatment with ibudilast allowed for deeper extinction of active lever pressing. Quantitative mass spectrometry based proteomics on the PFC identified one potential hit; the synaptic signaling protein phosphatidylethanolamine-binding protein 1 (PEBP1). While methamphetamine intake was associated with reduced PEBP1 protein levels, treatment with ibudilast reversed this effect. Furthermore, decreased PEBP1 expression was correlated with subsequent activation of Raf-1, MEK, and ERK signaling components of the mitogen-activated protein kinase cascade (MAPK). Raf-1, MEK, and ERK expression levels were also attenuated by ibudilast treatment. CONCLUSION PEBP1, given its synaptic localization and its role as a signaling molecule acting via the ERK/MAPK pathway, could be a potential therapeutic target mediating drug-seeking behaviors associated with neuroinflammation.
Collapse
|
46
|
Poupon L, Kerckhove N, Vein J, Lamoine S, Authier N, Busserolles J, Balayssac D. Minimizing chemotherapy-induced peripheral neuropathy: preclinical and clinical development of new perspectives. Expert Opin Drug Saf 2015; 14:1269-82. [DOI: 10.1517/14740338.2015.1056777] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Giron SE, Griffis CA, Burkard JF. Chronic Pain and Decreased Opioid Efficacy: An Inflammatory Link. Pain Manag Nurs 2015; 16:819-31. [PMID: 25962543 DOI: 10.1016/j.pmn.2015.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/16/2015] [Accepted: 04/02/2015] [Indexed: 01/08/2023]
Abstract
Chronic pain is a devastating amalgam of symptoms that affects millions of Americans at tremendous cost to our healthcare system and, more importantly, to patients' quality of life. Literature and research demonstrate that neuroimmune cells called glia are not only responsible for initiating and maintaining part of the chronic pain disease process, but also release inflammatory molecules responsible for decreasing the efficacy of one of the most prominent treatments for pain, opioid analgesia. This article describes chronic pain as a disease process that has ineffective treatment modalities, explores the mechanisms of glial cell activation and inflammatory responses that lead to chronic pain and decreased opioid treatment efficacy, and hypothesizes novel chronic pain treatment modalities based on the glial cell inactivation and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Sarah E Giron
- Department of Anesthesiology, University of Southern California Keck School of Medicine, Los Angeles, California.
| | - Charles A Griffis
- Department of Anesthesiology, University of California at Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Joseph F Burkard
- University of San Diego Hahn School of Nursing and Health Science, San Diego, California
| |
Collapse
|
48
|
Brennan TV, Rendell VR, Yang Y. Innate immune activation by tissue injury and cell death in the setting of hematopoietic stem cell transplantation. Front Immunol 2015; 6:101. [PMID: 25852683 PMCID: PMC4360715 DOI: 10.3389/fimmu.2015.00101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/23/2015] [Indexed: 11/22/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) with donor lymphocyte infusion is the mainstay of treatment for many types of hematological malignancies, but the therapeutic effect and prevention of relapse is complicated by donor T-cell recognition and attack of host tissue in a process known as graft-versus-host disease (GvHD). Cytotoxic myeloablative conditioning regimens used prior to Allo-HSCT result in the release of endogenous innate immune activators that are increasingly recognized for their role in creating a pro-inflammatory milieu. This increased inflammatory state promotes allogeneic T-cell activation and the induction and perpetuation of GvHD. Here, we review the processes of cellular response to injury and cell death that are relevant following Allo-HSCT and present the current evidence for a causative role of a variety of endogenous innate immune activators in the mediation of sterile inflammation following Allo-HSCT. Finally, we discuss the potential therapeutic strategies that target the endogenous pathways of innate immune activation to decrease the incidence and severity of GvHD following Allo-HSCT.
Collapse
Affiliation(s)
- Todd V Brennan
- Department of Surgery, Duke University , Durham, NC , USA
| | | | - Yiping Yang
- Department of Medicine, Duke University , Durham, NC , USA ; Department of Immunology, Duke University , Durham, NC , USA
| |
Collapse
|
49
|
Alfonso Romero-Sandoval E, Sweitzer S. Nonneuronal central mechanisms of pain: glia and immune response. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:325-58. [PMID: 25744678 DOI: 10.1016/bs.pmbts.2014.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of central glial cells in the mechanisms underlying pain has been intensively studied in the last two decades. Most studies on glia and pain focused on the potential detrimental role of glial cells following noxious stimulus/insults manifested as an "activation" or a "reactive" state (increase in glial marker expression and production of proinflammatory/nociceptive molecules). Therefore, "activated" or "reactive" glial cells became a target for the future generation of drugs to treat chronic pain. Several glial modulators that reduce the activation of glial cells have shown great efficacy in multiple animal (rodents mostly) models of pain (acute, subacute, chronic, inflammatory, neuropathic, surgical, etc.). These encouraging findings inspired clinical trials that have been completed in the last 5 years. Unfortunately, all clinical trials with these glial modulators have failed to demonstrate efficacy for the treatment of pain. New lines of investigation and elegant experimental designs are shedding light on alternative glial functions, which demonstrate that "glial reactivity" is not necessarily deleterious in some pathological conditions. New strategies to validate findings through our current animal models are necessary to enhance the translational value of our preclinical studies. Also, more studies using human subjects would enhance our understanding of glial cells in the context of pain. This chapter explores the available literature to objectively ponder the potential role of glial cells in human pain conditions.
Collapse
Affiliation(s)
- E Alfonso Romero-Sandoval
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA.
| | - Sarah Sweitzer
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA
| |
Collapse
|
50
|
Abstract
Migraine and cluster headache are primary headache disorders commonly encountered in clinical practice. Despite the profound disability caused by these primary headache disorders, available acute and preventive treatment options are limited. Recent understanding of headache pathophysiology has led to the development of new drug formulations and novel drug targets that are extremely promising. This article will highlight several of the new treatments that are currently under investigation including novel delivery mechanisms of already existing medications, calcitonin gene-related peptide (CGRP) receptor antagonists, antibodies to CGRP and its receptor, serotonin receptor agonists, transient receptor potential vanilloid receptor modulators, orexin receptor antagonists, glial cell modulators, and neuromodulation. If data is supportive, these therapies will be welcome additions to the headache specialist's armamentarium.
Collapse
|