1
|
Acuña AM, Park C, Leyrer-Jackson JM, Olive MF. Promising immunomodulators for management of substance and alcohol use disorders. Expert Opin Pharmacother 2024; 25:867-884. [PMID: 38803314 PMCID: PMC11216154 DOI: 10.1080/14656566.2024.2360653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION The neuroimmune system has emerged as a novel target for the treatment of substance use disorders (SUDs), with immunomodulation producing encouraging therapeutic benefits in both preclinical and clinical settings. AREAS COVERED In this review, we describe the mechanism of action and immune response to methamphetamine, opioids, cocaine, and alcohol. We then discuss off-label use of immunomodulators as adjunctive therapeutics in the treatment of neuropsychiatric disorders, demonstrating their potential efficacy in affective and behavioral disorders. We then discuss in detail the mechanism of action and recent findings regarding the use of ibudilast, minocycline, probenecid, dexmedetomidine, pioglitazone, and cannabidiol to treat (SUDs). These immunomodulators are currently being investigated in clinical trials described herein, specifically for their potential to decrease substance use, withdrawal severity, central and peripheral inflammation, comorbid neuropsychiatric disorder symptomology, as well as their ability to improve cognitive outcomes. EXPERT OPINION We argue that although mixed, findings from recent preclinical and clinical studies underscore the potential benefit of immunomodulation in the treatment of the behavioral, cognitive, and inflammatory processes that underlie compulsive substance use.
Collapse
Affiliation(s)
- Amanda M. Acuña
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| | - Connor Park
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - Jonna M. Leyrer-Jackson
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - M. Foster Olive
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
2
|
Yin X, Gao Q, Li C, Yang Q, HongliangDong, Li Z. Leonurine alleviates vancomycin nephrotoxicity via activating PPARγ and inhibiting the TLR4/NF-κB/TNF-α pathway. Int Immunopharmacol 2024; 131:111898. [PMID: 38513573 DOI: 10.1016/j.intimp.2024.111898] [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: 02/04/2024] [Revised: 03/10/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Vancomycin (VCM) is the first-line antibiotic for severe infections, but nephrotoxicity limits its use. Leonurine (Leo) has shown protective effects against kidney damage. However, the effect and mechanism of Leo on VCM nephrotoxicity remain unclear. In this study, mice and HK-2 cells exposed to VCM were treated with Leo. Biochemical and pathological analysis and fluorescence probe methods were performed to examine the role of Leo in VCM nephrotoxicity. Immunohistochemistry, q-PCR, western blot, FACS, and Autodock software were used to verify the mechanism. The present results indicate that Leo significantly alleviates VCM-induced renal injury, morphological damage, and oxidative stress. Increased intracellular and mitochondrial ROS in HK-2 cells and decreased mitochondrial numbers in mouse renal tubular epithelial cells were reversed in Leo-administrated groups. In addition, molecular docking analysis using Autodock software revealed that Leo binds to the PPARγ protein with high affinity. Mechanistic exploration indicated that Leo inhibited VCM nephrotoxicity via activating PPARγ and inhibiting the TLR4/NF-κB/TNF-α inflammation pathway. Taken together, our results indicate that the PPARγ inhibition and inflammation reactions were implicated in the VCM nephrotoxicity and provide a promising therapeutic strategy for renal injury.
Collapse
Affiliation(s)
- Xuedong Yin
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Chensuizi Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Medicine, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Qiaoling Yang
- Department of Pharmacy, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - HongliangDong
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200120, China.
| | - Zhiling Li
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
3
|
Tahamtan M, Aghaei I, Shabani M, Nazari A, Pooladvand V, Razavinasab M. Peroxisome proliferator-activated receptor-γ doesn't modify altered electrophysiological properties of the CA1 pyramidal neurons in a rat model of hepatic cirrhosis. Metab Brain Dis 2022; 37:2687-2697. [PMID: 35943675 DOI: 10.1007/s11011-022-01057-7] [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: 05/29/2022] [Accepted: 07/22/2022] [Indexed: 10/15/2022]
Abstract
Regarding the low quality of life due to the cognitive complications in the patients with hepatic cirrhosis (HC), the goal of this study was to examine the possible neuroprotective effect of pioglitazone (PIO) on the electrophysiological alterations of hippocampus, a major area of cognition, in the experimental model of bile duct ligation (BDL). We used adult male Wistar rats in the present study to perform BDL or sham surgery. Pioglitazone was administered in BDL rats two weeks after the surgery for the next continuous four weeks. The effects of pioglitazone on BDL-induced electrophysiological alterations of the CA1 pyramidal neurons in the hippocampus were evaluated by whole-cell patch clamp recordings. Our findings demonstrated that chronic administration of PIO could not reverse the electrophysiological changes in the CA1 pyramidal neurons of the hippocampus in BDL rats but could improve the hepatic dysfunction.Together, the results of this study suggest that PIO administration cannot counteract altered intrinsic properties of the hippocampal neurons which has been shown recently as an involved mechanism of the cognitive impairments in hepatic encephalopathy (HE).
Collapse
Affiliation(s)
- Mahshid Tahamtan
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iraj Aghaei
- Neuroscience Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
| | - Abbas Nazari
- Department of Biology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Vahid Pooladvand
- Biochemical Department, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Moazamehosadat Razavinasab
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, 76198-13159, Kerman, Iran.
| |
Collapse
|
4
|
Nakagawa Y, Lee J, Liu Y, Abbasi S, Hong T, Cabral H, Uchida S, Ebara M. Microglial Immunoregulation by Apoptotic Cellular Membrane Mimetic Polymeric Particles. ACS Macro Lett 2022; 11:270-275. [PMID: 35574780 DOI: 10.1021/acsmacrolett.1c00643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Phosphatidylserine (PtdSer), one of the phospholipids that the apoptotic cell exposes, has emerged for anti-inflammatory therapy via polarizing inflammatory microglia (Mi1) to anti-inflammatory phenotype (Mi2). In this study, we report microglia polarization effect of PtdSer-exposing polymeric particles (PSPs). PSPs upregulated Mi2 microglia and suppressed Mi1 microglia through peroxisome proliferator-activated receptor gamma upregulation in vitro and in vivo. This study highlights the potential of PSPs for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Yasuhiro Nakagawa
- School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo 152-8550, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1, Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Jeonggyu Lee
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1, Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yihua Liu
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1, Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
| | - Taehun Hong
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Horacio Cabral
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kanagawa 210-0821, Japan
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Graduate School of Medicine, Kyoto Prefectural University of Medicine, 1-5, Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Mitsuhiro Ebara
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), 1-1, Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Graduate School of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 162-8601, Japan
| |
Collapse
|
5
|
Rahman S, Rahman ZI, Ronan PJ, Lutfy K, Bell RL. Adolescent opioid abuse: Role of glial and neuroimmune mechanisms. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:147-165. [PMID: 34801168 DOI: 10.1016/bs.irn.2021.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Opioids are widely prescribed for pain management, and prescription opioid misuse in adolescents has become a major epidemic in the United States and worldwide. Emerging data indicate that adolescence represents a critical period of brain development, and exposure to opioids during adolescence may increase the risk of addiction in adulthood. There is growing evidence that disruptions in brain glial function may be implicated in numerous chronic neuropathologies. Evidence suggests that glial mechanisms have an important role in the development and maintenance of opioid abuse and the risk for addiction. This review will describe glial and neuroimmune mechanisms involved in opioid use disorders during adolescence, which may increase substance use disorder liability later in life. Moreover, this review will identify some important neuro-glial targets, involved in opioid abuse and addiction, to develop future preventions and treatment strategies.
Collapse
Affiliation(s)
- S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States.
| | - Z I Rahman
- University of Minnesota Medical School, Minneapolis, MN, United States
| | - P J Ronan
- Department of Psychiatry and Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States; Research Service, Sioux Falls VA Healthcare System, Sioux Falls, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - R L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
6
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
7
|
Kumar N, Sharma N, Khera R, Gupta R, Mehan S. Guggulsterone ameliorates ethidium bromide-induced experimental model of multiple sclerosis via restoration of behavioral, molecular, neurochemical and morphological alterations in rat brain. Metab Brain Dis 2021; 36:911-925. [PMID: 33635478 DOI: 10.1007/s11011-021-00691-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/11/2021] [Indexed: 11/30/2022]
Abstract
Multiple Sclerosis (MS) is a progressive neurodegenerative disease with clinical signs of neuroinflammation and the central nervous system's demyelination. Numerous studies have identified the role of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) overexpression and the low level of peroxisome proliferator-activated receptor-gamma (PPAR-γ) in MS pathogenesis. Guggulsterone (GST), an active component derived from 'Commiphora Mukul,' has been used to treat various diseases. Traditional uses indicate that GST is a suitable agent for anti-inflammatory action. Therefore, we assessed the therapeutic potential of GST (30 and 60 mg/kg) in ethidium bromide (EB) induced demyelination in experimental rats and investigated the molecular mechanism by modulating the JAK/STAT and PPAR-γ receptor signaling. Wistar rats were randomly divided into six groups (n = 6). EB (0.1%/10 μl) was injected selectively in the intracerebropeduncle (ICP) region for seven days to cause MS-like manifestations. The present study reveals that long-term administration of GST for 28 days has a neuroprotective effect by improving behavioral deficits (spatial cognition memory, grip, and motor coordination) associated with lower STAT-3 levels. While elevating PPAR-γ and myelin basic protein levels in rat brains are consistent with the functioning of both signaling pathways. Also, GST modulates the neurotransmitter level by increasing Ach, dopamine, serotonin and by reducing glutamate. Moreover, GST ameliorates inflammatory cytokines (TNF, IL-1β), and oxidative stress markers (AchE, SOD, catalase, MDA, GSH, nitrite). In addition, GST prevented apoptosis, as demonstrated by the reduction of caspase-3 and Bax. Simultaneously, Bcl-2 elevation and the restoration of gross morphology alterations are also recovered by long-term GST treatment. Therefore, it can be concluded that GST may be a potential alternative drug candidate for MS-related motor neuron dysfunctions.
Collapse
Affiliation(s)
- Nitish Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Nidhi Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Rishabh Khera
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ria Gupta
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
8
|
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
|
9
|
Glial neuroimmune signaling in opioid reward. Brain Res Bull 2019; 155:102-111. [PMID: 31790721 DOI: 10.1016/j.brainresbull.2019.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
The opioid epidemic is a growing public concern affecting millions of people worldwide. Opioid-induced reward is the initial and key process leading to opioid abuse and addiction. Therefore, a better understanding of opioid reward may be helpful in developing a treatment for opioid addiction. Emerging evidence suggests that glial cells, particularly microglia and astrocytes, play an essential role in modulating opioid reward. Indeed, glial cells and their associated immune signaling actively regulate neural activity and plasticity, and directly modulate opioid-induced rewarding behaviors. In this review, we describe the neuroimmune mechanisms of how glial cells affect synaptic transmission and plasticity as well as how opioids can activate glial cells affecting the glial-neuronal interaction. Last, we summarize current attempts of applying glial modulators in treating opioid reward.
Collapse
|
10
|
d'Angelo M, Castelli V, Catanesi M, Antonosante A, Dominguez-Benot R, Ippoliti R, Benedetti E, Cimini A. PPARγ and Cognitive Performance. Int J Mol Sci 2019; 20:ijms20205068. [PMID: 31614739 PMCID: PMC6834178 DOI: 10.3390/ijms20205068] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Recent findings have led to the discovery of many signaling pathways that link nuclear receptors with human conditions, including mental decline and neurodegenerative diseases. PPARγ agonists have been indicated as neuroprotective agents, supporting synaptic plasticity and neurite outgrowth. For these reasons, many PPARγ ligands have been proposed for the improvement of cognitive performance in different pathological conditions. In this review, the research on this issue is extensively discussed.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Reyes Dominguez-Benot
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
11
|
Kohno M, Link J, Dennis LE, McCready H, Huckans M, Hoffman WF, Loftis JM. Neuroinflammation in addiction: A review of neuroimaging studies and potential immunotherapies. Pharmacol Biochem Behav 2019; 179:34-42. [PMID: 30695700 DOI: 10.1016/j.pbb.2019.01.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Addiction is a worldwide public health problem and this article reviews scientific advances in identifying the role of neuroinflammation in the genesis, maintenance, and treatment of substance use disorders. With an emphasis on neuroimaging techniques, this review examines human studies of addiction using positron emission tomography to identify binding of translocator protein (TSPO), which is upregulated in reactive glial cells and activated microglia during pathological states. High TSPO levels have been shown in methamphetamine use but exhibits variable patterns in cocaine use. Alcohol and nicotine use, however, are associated with lower TSPO levels. We discuss how mechanistic differences at the neurotransmitter and circuit level in the neural effects of these agents and subsequent immune response may explain these observations. Finally, we review the potential of anti-inflammatory drugs, including ibudilast, minocycline, and pioglitazone, to ameliorate the behavioral and cognitive consequences of addiction.
Collapse
Affiliation(s)
- Milky Kohno
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jeanne Link
- Center for Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - Laura E Dennis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Holly McCready
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Marilyn Huckans
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - William F Hoffman
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
12
|
Jones JD, Bisaga A, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, Madera G, Doernberg M, Comer SD. The PPARγ Agonist Pioglitazone Fails to Alter the Abuse Potential of Heroin, But Does Reduce Heroin Craving and Anxiety. J Psychoactive Drugs 2018; 50:390-401. [PMID: 30204554 DOI: 10.1080/02791072.2018.1508789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Possibly through its effects on glia, the peroxisome proliferator-activated gamma receptor (PPARγ) agonist pioglitazone (PIO) has been shown to alter the effects of heroin in preclinical models. Until now, these results have not been assessed in humans. Heroin-dependent participants were randomized to either active (45 mg, n = 14) or placebo (0 mg, n = 16) PIO maintenance for the duration of the three-week study. After stabilization on buprenorphine (8 mg), participants began a two-week testing period. On the first to fourth test days, participants could self-administer drug or money by making verbal choices for either option. On the fifth day, active heroin and money were administered and participants could work to receive heroin or money using a progressive ratio choice procedure. Test days 6-10 were identical to test days 1-5 with the exception that, during one of the test weeks, placebo was available on the first four days, and during the other week heroin was available. PIO failed to alter the reinforcing or positive subjective effects of heroin, but it did reduce heroin craving and overall anxiety. Although we were unable to replicate the robust effects found in preclinical models, these data provide an indication of drug effects that deserves further exploration.
Collapse
Affiliation(s)
- Jermaine D Jones
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Adam Bisaga
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Verena E Metz
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Jeanne M Manubay
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Shanthi Mogali
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Roberto Ciccocioppo
- b Department of Experimental Medicine and Public Health, School of Pharmacy, Pharmacology Unit , University of Camerino , Macerata , Italy
| | - Gabriela Madera
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Molly Doernberg
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| | - Sandra D Comer
- a Division on Substance Use Disorders , New York State Psychiatric Institute and College of Physicians and Surgeons of Columbia University , New York , NY , USA
| |
Collapse
|
13
|
Zhang L, Gao J, Tang P, Chong L, Liu Y, Liu P, Zhang X, Chen L, Hou C. Nuciferine inhibits LPS-induced inflammatory response in BV2 cells by activating PPAR-γ. Int Immunopharmacol 2018; 63:9-13. [PMID: 30056259 DOI: 10.1016/j.intimp.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/01/2018] [Accepted: 07/16/2018] [Indexed: 01/08/2023]
Abstract
Nuciferine, a bioactive component extracted from the lotus leaf, has been reported to have various anti-inflammatory effects. In the present study, we aimed to investigate the anti-inflammatory effects and mechanism of nuciferine on lipopolysaccharide (LPS)-stimulated BV2 microglia cells. The anti-inflammatory activity of nuciferine was measured by ELISA to detect the inflammatory mrdiators secretion in LPS-simulated BV2 microglia cells. The results demonstrated that nuciferine significantly inhibited LPS-induced TNF-α, IL-1β, PGE2 and NO secretion. LPS-induced NF-κB activation was also suppressed by nuciferine. Further studies showed that nuciferine increased the expression of PPAR-γ. Functional aspects were analyzed using PPAR-γ specific inhibitor GW9662, which attenuated the LPS-induced secretion of proinflammatory mediators, such as TNF-α, IL-1β, PGE2, and NO. In conclusion, these results suggested that nuciferine activated PPAR-γ, which subsequently inhibited LPS-induced inflammation in BV2 cells.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Jinghua Gao
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Peng Tang
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Li Chong
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Yue Liu
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Peng Liu
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Xin Zhang
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Li Chen
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Chen Hou
- Department of Neurology, Shaanxi Provincial People's Hospital, and the Third Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China.
| |
Collapse
|
14
|
Jones JD, Comer SD, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, Martinez S, Mumtaz M, Bisaga A. Pioglitazone, a PPARγ agonist, reduces nicotine craving in humans, with marginal effects on abuse potential. Pharmacol Biochem Behav 2017; 163:90-100. [PMID: 29020601 PMCID: PMC5959043 DOI: 10.1016/j.pbb.2017.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/20/2017] [Accepted: 10/04/2017] [Indexed: 11/26/2022]
Abstract
Possibly through their actions upon glia, peroxisome proliferator-activated receptor agonists (PPAR) have been shown to alter the abuse potential of addictive drugs in several preclinical models. The current study extends this research into the human laboratory as the first clinical study into the effects of the PPAR gamma agonist, pioglitazone, on the abuse potential of nicotine. Heavy smokers were recruited for this 3-week study. Upon admission, participants were randomized to either active (45mg, n=14) or placebo (0mg, n=13) PIO maintenance conditions for the duration of the study. After 5-7days of stabilization on a 7mg nicotine patch, participants began laboratory testing. On the 1st-4th test days, participants could self-administer cigarettes or receive money by making verbal choices for either option. On the 5th day, participants were administered 10 puffs of their usual brand of cigarette in the morning and later chose between smoking and money by making finger presses on a computer mouse in a progressive ratio self-administration task. Later on the 5th day participants also underwent a smoking cue exposure session. The 8th-11th test days were identical to the 1st-4th test days with the exception that during one of the test weeks de-nicotinized cigarettes were available, and during the other nicotinized cigarettes were available. Nicotinized cigarettes were always administered on the 5th and 12th days. On some measures PIO increased indicators of abuse potential, though this effect was typically not statistically significant. However, PIO did significantly reduce measures of craving.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Corresponding author. (J.D. Jones)
| | - Sandra D. Comer
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Verena E. Metz
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Jeanne M. Manubay
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Shanthi Mogali
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, Macerata 62032, Italy
| | - Suky Martinez
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Translational Research Training Program in Addiction, City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Mudassir Mumtaz
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA,Translational Research Training Program in Addiction, City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Adam Bisaga
- Division on Substance Use Disorders, Department of Psychiatry, New York State Psychiatric Institute, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| |
Collapse
|
15
|
Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Front Immunol 2017; 8:1520. [PMID: 29176977 PMCID: PMC5686097 DOI: 10.3389/fimmu.2017.01520] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/26/2017] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia play crucial roles during central nervous system development, homeostasis and acute events such as infection or injury. The diverse functions of tissue macrophages and microglia are mirrored by equally diverse phenotypes. A model of inflammatory/M1 versus a resolution phase/M2 macrophages has been widely used. However, the complexity of macrophage function can only be achieved by the existence of varied, plastic and tridimensional macrophage phenotypes. Understanding how tissue macrophages integrate environmental signals via molecular programs to define pathogen/injury inflammatory responses provides an opportunity to better understand the multilayered nature of macrophages, as well as target and modulate cellular programs to control excessive inflammation. This is particularly important in MS and other neuroinflammatory diseases, where chronic inflammatory macrophage and microglial responses may contribute to pathology. Here, we perform a comprehensive review of our current understanding of how molecular pathways modulate tissue macrophage phenotype, covering both classic pathways and the emerging role of microRNAs, receptor-tyrosine kinases and metabolism in macrophage phenotype. In addition, we discuss pathway parallels in microglia, novel markers helpful in the identification of peripheral macrophages versus microglia and markers linked to their phenotype.
Collapse
Affiliation(s)
- Stephanie A Amici
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Joycelyn Dong
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,McCormick School of Engineering, Division of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
16
|
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend 2017; 180:156-170. [PMID: 28892721 PMCID: PMC5790191 DOI: 10.1016/j.drugalcdep.2017.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The plenary session at the 2016 Behavior, Biology and Chemistry: Translational Research in Addiction Conference focused on glia as potential players in the development, persistence and treatment of substance use disorders. Glia partake in various functions that are important for healthy brain activity. Drugs of abuse alter glial cell activity producing several perturbations in brain function that are thought to contribute to behavioral changes associated with substance use disorders. Consequently, drug-induced changes in glia-driven processes in the brain represent potential targets for pharmacotherapeutics treating substance use disorders. METHODS Four speakers presented preclinical and clinical research illustrating the effects that glial modulators have on abuse-related behavioral effects of psychostimulants and opioids. This review highlights some of these findings and expands its focus to include other research focused on drug-induced glia abnormalities and glia-focused treatment approaches in substance use disorders. RESULTS Preclinical findings show that drugs of abuse induce neuroinflammatory signals and disrupt glutamate homeostasis through their interaction with microglia and astrocytes. Preclinical and clinical studies testing the effects of glial modulators show general effectiveness in reducing behaviors associated with substance use disorders. CONCLUSIONS The contribution of drug-induced glial activity continues to emerge as an intriguing target for substance use disorder treatments. Clinical investigations of glial modulators have yielded promising results on substance use measures and indicate that they are generally safe and well-tolerated. However, results have not been entirely positive and more questions remain for continued exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Ryan K. Bachtell
- Department of Psychology and Neuroscience, and Center for Neuroscience, UCB 345, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Keith G. Heinzerling
- Department of Family Medicine and Center for Behavioral and Addiction Medicine, UCLA, Los Angeles, CA, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
17
|
Protective Effect of PPAR γ Agonists on Cerebellar Tissues Oxidative Damage in Hypothyroid Rats. Neurol Res Int 2016; 2016:1952561. [PMID: 28116157 PMCID: PMC5220477 DOI: 10.1155/2016/1952561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/06/2016] [Accepted: 11/30/2016] [Indexed: 12/20/2022] Open
Abstract
The aim of the current study was to investigate the effects of peroxisome proliferator-activated receptor gamma (PPARγ) agonists on cerebellar tissues oxidative damage in hypothyroid rats. The animals included seven groups: group I (control), the animals received drinking water; group II, the animals received 0.05% propylthiouracil (PTU) in drinking water; besides PTU, the animals in groups III, IV, V, VI, and VII, were injected with 20 mg/kg vitamin E (Vit E), 10 or 20 mg/kg pioglitazone, and 2 or 4 mg/kg rosiglitazone, respectively. The animals were deeply anesthetized and the cerebellar tissues were removed for biochemical measurements. PTU administration reduced thiol content, superoxide dismutase (SOD), and catalase (CAT) activities in the cerebellar tissues while increasing malondialdehyde (MDA) and nitric oxide (NO) metabolites. Vit E, pioglitazone, and rosiglitazone increased thiol, SOD, and CAT in the cerebellar tissues while reducing MDA and NO metabolites. The results of present study showed that, similar to Vit E, both rosiglitazone and pioglitazone as PPARγ agonists exerted protective effects against cerebellar tissues oxidative damage in hypothyroid rats.
Collapse
|
18
|
Vallée A, Lecarpentier Y. Alzheimer Disease: Crosstalk between the Canonical Wnt/Beta-Catenin Pathway and PPARs Alpha and Gamma. Front Neurosci 2016; 10:459. [PMID: 27807401 PMCID: PMC5069291 DOI: 10.3389/fnins.2016.00459] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
The molecular mechanisms underlying the pathophysiology of Alzheimer's disease (AD) are still not fully understood. In AD, Wnt/beta-catenin signaling has been shown to be downregulated while the peroxisome proliferator-activated receptor (PPAR) gamma (mARN and protein) is upregulated. Certain neurodegenerative diseases share the same Wnt/beta-catenin/PPAR gamma profile, such as bipolar disorder and schizophrenia. Conversely, other NDs share an opposite profile, such as amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, multiple sclerosis, and Friedreich's ataxia. AD is characterized by the deposition of extracellular Abeta plaques and the formation of intracellular neurofibrillary tangles in the central nervous system (CNS). Activation of Wnt signaling or inhibition of both glycogen synthase kinase-3beta and Dickkopf 1, two key negative regulators of the canonical Wnt pathway, are able to protect against Abeta neurotoxicity and to ameliorate cognitive performance in AD patients. Although PPAR gamma is upregulated in AD patients, and despite the fact that it has been shown that the PPAR gamma and Wnt/beta catenin pathway systems work in an opposite manner, PPAR gamma agonists diminish learning and memory deficits, decrease Abeta activation of microglia, and prevent hippocampal and cortical neurons from dying. These beneficial effects observed in AD transgenic mice and patients might be partially due to the anti-inflammatory properties of PPAR gamma agonists. Moreover, activation of PPAR alpha upregulates transcription of the alpha-secretase gene and represents a new therapeutic treatment for AD. This review focuses largely on the behavior of two opposing pathways in AD, namely Wnt/beta-catenin signaling and PPAR gamma. It is hoped that this approach may help to develop novel AD therapeutic strategies integrating PPAR alpha signaling.
Collapse
Affiliation(s)
- Alexandre Vallée
- CHU Amiens Picardie, Université Picardie Jules VerneAmiens, France
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of PoitiersPoitiers, France
- AP-HP, Epidemiology and Clinical Research Department, University Hospital Bichat-Claude BernardParis, France
| | | |
Collapse
|
19
|
Rolland M, Li X, Sellier Y, Martin H, Perez-Berezo T, Rauwel B, Benchoua A, Bessières B, Aziza J, Cenac N, Luo M, Casper C, Peschanski M, Gonzalez-Dunia D, Leruez-Ville M, Davrinche C, Chavanas S. PPARγ Is Activated during Congenital Cytomegalovirus Infection and Inhibits Neuronogenesis from Human Neural Stem Cells. PLoS Pathog 2016; 12:e1005547. [PMID: 27078877 PMCID: PMC4831785 DOI: 10.1371/journal.ppat.1005547] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/12/2016] [Indexed: 11/25/2022] Open
Abstract
Congenital infection by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae of the central nervous system, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities (0.1% of all births). To gain insight on the impact of HCMV on neuronal development, we used both neural stem cells from human embryonic stem cells (NSC) and brain sections from infected fetuses and investigated the outcomes of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor critical in the developing brain. We observed that HCMV infection dramatically impaired the rate of neuronogenesis and strongly increased PPARγ levels and activity. Consistent with these findings, levels of 9-hydroxyoctadecadienoic acid (9-HODE), a known PPARγ agonist, were significantly increased in infected NSCs. Likewise, exposure of uninfected NSCs to 9-HODE recapitulated the effect of infection on PPARγ activity. It also increased the rate of cells expressing the IE antigen in HCMV-infected NSCs. Further, we demonstrated that (1) pharmacological activation of ectopically expressed PPARγ was sufficient to induce impaired neuronogenesis of uninfected NSCs, (2) treatment of uninfected NSCs with 9-HODE impaired NSC differentiation and (3) treatment of HCMV-infected NSCs with the PPARγ inhibitor T0070907 restored a normal rate of differentiation. The role of PPARγ in the disease phenotype was strongly supported by the immunodetection of nuclear PPARγ in brain germinative zones of congenitally infected fetuses (N = 20), but not in control samples. Altogether, our findings reveal a key role for PPARγ in neurogenesis and in the pathophysiology of HCMV congenital infection. They also pave the way to the identification of PPARγ gene targets in the infected brain. Congenital infection by human cytomegalovirus (HCMV) might result in permanent neurological sequelae, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities. Infants with such sequelae represent about 0.1% of all live births (>5500 per year in the USA). Given the considerable health and societal burden, a better insight on disease pathogenesis is urgently needed to design new therapeutic or prognostic tools. Here, we studied the impact of HCMV on neuronal development, using human neural progenitors (NSC) as a disease model. In particular, we investigated the outcome of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ, a key protein in the regulation of metabolism, inflammation and cell differentiation. We disclosed that HCMV infection strongly increases levels and activity of PPARγ in NSCs. In vitro experiments showed that PPARγ activity inhibits the differentiation of NSCs into neurons. We also found increased PPARγ expression in brains of in utero infected fetuses, but not in controls, suggesting that PPARγ is a key effector of HCMV infection also in vivo. Our study provides new insights on the pathogenesis of HCMV infection and paves the way to the discovery of PPARγ-related molecules secreted in the infected brain.
Collapse
Affiliation(s)
- Maude Rolland
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Xiaojun Li
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yann Sellier
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hélène Martin
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Teresa Perez-Berezo
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Benjamin Rauwel
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | | | - Bettina Bessières
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jacqueline Aziza
- Département d'Anatomie Pathologique, IUCT-Oncopole, Toulouse, France
| | - Nicolas Cenac
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Minhua Luo
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Charlotte Casper
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- Neonatal Unit, Children’s Hospital, Toulouse, France
| | - Marc Peschanski
- I-STEM, INSERM U861, AFM, Evry, France
- CECS, UEVE U861, Evry, France
| | - Daniel Gonzalez-Dunia
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Marianne Leruez-Ville
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Christian Davrinche
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Stéphane Chavanas
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- * E-mail:
| |
Collapse
|
20
|
Morales-Martínez A, Sánchez-Mendoza A, Martínez-Lazcano JC, Pineda-Farías JB, Montes S, El-Hafidi M, Martínez-Gopar PE, Tristán-López L, Pérez-Neri I, Zamorano-Carrillo A, Castro N, Ríos C, Pérez-Severiano F. Essential fatty acid-rich diets protect against striatal oxidative damage induced by quinolinic acid in rats. Nutr Neurosci 2016; 20:388-395. [DOI: 10.1080/1028415x.2016.1147683] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Adriana Morales-Martínez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
- Laboratorio de Investigación de Bioquímica y Biofísica Computacional, Escuela Nacional de Medicina y de Homeopatía del IPN, Guillermo Massieu H. #239. La Escalera, Gustavo A. Madero, 07320 México D.F., México
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano #1, Col. Sección XVI, Tlalpan, México D.F., México
| | - Juan Carlos Martínez-Lazcano
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Jorge Baruch Pineda-Farías
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Mohammed El-Hafidi
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Col. Sección XVI. Tlalpan, México D.F., México
| | - Pablo Eliasib Martínez-Gopar
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Luis Tristán-López
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Iván Pérez-Neri
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Absalom Zamorano-Carrillo
- Laboratorio de Investigación de Bioquímica y Biofísica Computacional, Escuela Nacional de Medicina y de Homeopatía del IPN, Guillermo Massieu H. #239. La Escalera, Gustavo A. Madero, 07320 México D.F., México
| | - Nelly Castro
- Laboratorio de Neuropsicofarmacología, Instituto Nacional de Neurología y Neurocirugía, Insurgentes sur 3877, La Fama, Tlalpan, 14269 México D.F., México
| | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur #3877, Col. La Fama, Tlalpan, 14269 México D.F., México
| |
Collapse
|
21
|
Abstract
The molecular mechanism of neuronal loss and synaptic damage in Alzheimer's disease (AD), Parkinson's disease dementia (PDD), frontotemporal dementia (FTD) and Lewy body dementia (LBD) is poorly understood and could differ among different types of neurodegenerative processes. However, the presence of neuroinflammation is a common feature of dementia. In this setting, reactive microgliosis, oxidative damage and mitochondrial dysfunction are associated with the pathogenesis of all types of neurodegenerative dementia. Moreover, an increased body of evidence suggests that microglia may play a central role in AD progression. In this paper, we review the scientific literature on neuroinflammation related to the most common neurodegenerative dementias (AD, PDD, FTD and LBD) focussing on the possible molecular mechanisms and the available clinical evidence. Furthermore, we discuss the neuroimaging techniques that are currently used for the study of neuroinflammation in human brain.
Collapse
Affiliation(s)
- Giuseppe Pasqualetti
- Division of Brain Sciences, Department of Medicine, Imperial College London, 1st Floor B Block, Du Cane Road, London, W12 0NN, UK
| | | | | |
Collapse
|
22
|
Jones JD, Sullivan MA, Manubay JM, Mogali S, Metz VE, Ciccocioppo R, Comer SD. The effects of pioglitazone, a PPARγ receptor agonist, on the abuse liability of oxycodone among nondependent opioid users. Physiol Behav 2015; 159:33-9. [PMID: 26455893 DOI: 10.1016/j.physbeh.2015.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/31/2015] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
AIMS Activation of PPARγ by pioglitazone (PIO) has shown some efficacy in attenuating addictive-like responses in laboratory animals. The ability of PIO to alter the effects of opioids in humans has not been characterized in a controlled laboratory setting. The proposed investigation sought to examine the effects of PIO on the subjective, analgesic, physiological and cognitive effects of oxycodone (OXY). METHODS During this investigation, nondependent prescription opioid abusers (N=17 completers) were maintained for 2-3weeks on ascending daily doses of PIO (0mg, 15mg, 45mg) prior to completing a laboratory session assessing the aforementioned effects of OXY [using a within-session cumulative dosing procedure (0, 10, and 20mg, cumulative dose=30mg)]. RESULTS OXY produced typical mu opioid agonist effects: miosis, decreased pain perception, and decreased respiratory rate. OXY also produced dose-dependent increases in positive subjective responses. Yet, ratings such as: drug "liking," "high," and "good drug effect," were not significantly altered as a function of PIO maintenance dose. DISCUSSION These data suggest that PIO may not be useful for reducing the abuse liability of OXY. These data were obtained with a sample of nondependent opioid users and therefore may not be applicable to dependent populations or to other opioids. Although PIO failed to alter the abuse liability of OXY, the interaction between glia and opioid receptors is not well understood so the possibility remains that medications that interact with glia in other ways may show more promise.
Collapse
Affiliation(s)
- Jermaine D Jones
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA.
| | - Maria A Sullivan
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Jeanne M Manubay
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Shanthi Mogali
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Verena E Metz
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, Macerata 62032, Italy
| | - Sandra D Comer
- Division of Substance Abuse, New York Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, 1051 Riverside Drive, Unit 120, New York, NY 10032, USA
| |
Collapse
|
23
|
Protective effect of pioglitazone on morphine-induced neuroinflammation in the rat lumbar spinal cord. J Biomed Sci 2015; 22:82. [PMID: 26394827 PMCID: PMC4580127 DOI: 10.1186/s12929-015-0187-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 09/10/2015] [Indexed: 12/13/2022] Open
Abstract
Background Morphine-induced tolerance is associated with the spinal neuroinflammation. The aim of this study was to explore the effects of oral administration of the pioglitazone, the peroxisome proliferator activated receptor gamma (PPAR-γ) agonist, on the morphine-induced neuroinflammation in the lumbar region of the male Wistar rat spinal cord. Results Co-administration of the pioglitazone with morphine not only attenuated morphine-induced tolerance, but also prevented the up-regulation of pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin-1beta, and interleukin 6) and nuclear factor-kappa B activity. Administration of the GW-9662 antagonized the above mentioned effects of the pioglitazone. Conclusions It is concluded that oral administration of the pioglitazone attenuates morphine-induced tolerance and the neuroinflammation in the lumbar region of the rat spinal cord. This action of the pioglitazone may be, at least in part, due to an interaction with the spinal pro-inflammatory cytokine expression and the nuclear factor-kappa B activity.
Collapse
|
24
|
Kim JY, Kim N, Yenari MA. Mechanisms and potential therapeutic applications of microglial activation after brain injury. CNS Neurosci Ther 2014; 21:309-19. [PMID: 25475659 DOI: 10.1111/cns.12360] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 12/14/2022] Open
Abstract
As the resident immune cells of the central nervous system, microglia rapidly respond to brain insults, including stroke and traumatic brain injury. Microglial activation plays a major role in neuronal cell damage and death by releasing a variety of inflammatory and neurotoxic mediators. Their activation is an early response that may exacerbate brain injury and many other stressors, especially in the acute stages, but are also essential to brain recovery and repair. The full range of microglial activities is still not completely understood, but there is accumulating knowledge about their role following brain injury. We review recent progress related to the deleterious and beneficial effects of microglia in the setting of acute neurological insults and the current literature surrounding pharmacological interventions for intervention.
Collapse
Affiliation(s)
- Jong-Youl Kim
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
25
|
Ghavimi H, Hassanzadeh K, Maleki-Dizaji N, Azarfardian A, Ghasami S, Zolali E, Charkhpour M. Pioglitazone prevents morphine antinociception tolerance and withdrawal symptoms in rats. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:811-21. [DOI: 10.1007/s00210-014-0996-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 05/20/2014] [Indexed: 12/22/2022]
|
26
|
Liu S, Lin SJ, Li G, Kim E, Chen YT, Yang DR, Tan MHE, Yong EL, Chang C. Differential roles of PPARγ vs TR4 in prostate cancer and metabolic diseases. Endocr Relat Cancer 2014; 21:R279-300. [PMID: 24623743 DOI: 10.1530/erc-13-0529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ, NR1C3) and testicular receptor 4 nuclear receptor (TR4, NR2C2) are two members of the nuclear receptor (NR) superfamily that can be activated by several similar ligands/activators including polyunsaturated fatty acid metabolites, such as 13-hydroxyoctadecadienoic acid and 15-hydroxyeicosatetraenoic acid, as well as some anti-diabetic drugs such as thiazolidinediones (TZDs). However, the consequences of the transactivation of these ligands/activators via these two NRs are different, with at least three distinct phenotypes. First, activation of PPARγ increases insulin sensitivity yet activation of TR4 decreases insulin sensitivity. Second, PPARγ attenuates atherosclerosis but TR4 might increase the risk of atherosclerosis. Third, PPARγ suppresses prostate cancer (PCa) development and TR4 suppresses prostate carcinogenesis yet promotes PCa metastasis. Importantly, the deregulation of either PPARγ or TR4 in PCa alone might then alter the other receptor's influences on PCa progression. Knocking out PPARγ altered the ability of TR4 to promote prostate carcinogenesis and knocking down TR4 also resulted in TZD treatment promoting PCa development, indicating that both PPARγ and TR4 might coordinate with each other to regulate PCa initiation, and the loss of either one of them might switch the other one from a tumor suppressor to a tumor promoter. These results indicate that further and detailed studies of both receptors at the same time in the same cells/organs may help us to better dissect their distinct physiological roles and develop better drug(s) with fewer side effects to battle PPARγ- and TR4-related diseases including tumor and cardiovascular diseases as well as metabolic disorders.
Collapse
Affiliation(s)
- Su Liu
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Shin-Jen Lin
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Gonghui Li
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eungseok Kim
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Yei-Tsung Chen
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Dong-Rong Yang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - M H Eileen Tan
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Eu Leong Yong
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| | - Chawnshang Chang
- George Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, TaiwanGeorge Whipple Laboratory for Cancer ResearchDepartments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York 14642, USADepartment of Obstetrics and GynecologyNational University of Singapore, Singapore, SingaporeChawnshang Chang Liver Cancer Center and Department of UrologySir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, ChinaDepartment of Biological SciencesChonnam National University, Youngbong, Buk-Gu, Gwangju 500-757 KoreaCardiovascular Research InstituteNational University Health System and The Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, SingaporeSex Hormone Research CenterChina Medical University/Hospital, Taichung 404, Taiwan
| |
Collapse
|
27
|
Abstract
Alzheimer’s disease (AD) is complicated and difficult to fully understand, it might need multiple drug-discovery strategies to combat the disease. Regardless of the cause of AD, neuronal death in the brain plays a key role in AD progression and is directly linked to neuroinflammation. Thus, the regulation of neuroinflammatory processes might be a practical strategy for the treatment of AD. This review highlights the development of anti-neuroinflammatory agents that have shown promise in vitro or in vivo by attenuating microglial activation or cognitive decline. The agents are categorized based on the related signaling pathways, including the receptor for advanced glycation end products, p38 MAPKs, NF-κB and peroxisome proliferator-activated receptor γ; and inhibitors against microglial activation lacking clear mechanisms. These anti-neuroinflammatory agents support the concept and represent important chemical probes for the development of anti-neuroinflammatory drugs for the treatment of AD.
Collapse
|
28
|
Possible Involvement of PPAR-γ in the Anticonvulsant Effect of Aegle marmelos (L.) Correa. Neurochem Res 2013; 38:1624-31. [DOI: 10.1007/s11064-013-1064-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 04/24/2013] [Accepted: 04/26/2013] [Indexed: 12/11/2022]
|
29
|
Bérézowski V, Mysiorek C, Kuntz M, Pétrault O, Cecchelli R. [Dysfunction of the blood-brain barrier during ischaemia: a therapeutic concern]. Biol Aujourdhui 2012; 206:161-76. [PMID: 23171839 DOI: 10.1051/jbio/2012020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Indexed: 11/14/2022]
Abstract
Since it was discovered and its brain-protective role characterized, the blood-brain barrier (BBB), through the permeability-restricting action of the brain capillary endothelial cells, has been representing a hurdle for 95% of new medical compounds targeting the central nervous system. Recently, a BBB dysfunction is being found in an increasing number of pathologies such as brain ischaemic stroke, whose only therapy consists in a pharmacological thrombolysis limited to a small percentage of the admitted patients, because of the toxical effects of thrombolytics. And since the clinical failure of promising neuroprotectants, numerous studies of brain ischaemia were carried out, with physiopathological or pharmacological approaches refocused on the BBB, whose structural complexity is now expanded to perivascular cells, all forming a functional unit named the neurovascular unit (NVU). Nevertheless, in spite of the numerous molecular mechanisms identified, the process of BBB dysfunction in the ischaemia/reperfusion cascade remains insufficiently established to explain the pleiotropic action exerted by new pharmacological compounds, possibly protecting the entire NVU and representing potential treatments.
Collapse
|
30
|
Kanakasabai S, Pestereva E, Chearwae W, Gupta SK, Ansari S, Bright JJ. PPARγ agonists promote oligodendrocyte differentiation of neural stem cells by modulating stemness and differentiation genes. PLoS One 2012. [PMID: 23185633 PMCID: PMC3503969 DOI: 10.1371/journal.pone.0050500] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Neural stem cells (NSCs) are a small population of resident cells that can grow, migrate and differentiate into neuro-glial cells in the central nervous system (CNS). Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor transcription factor that regulates cell growth and differentiation. In this study we analyzed the influence of PPARγ agonists on neural stem cell growth and differentiation in culture. We found that in vitro culture of mouse NSCs in neurobasal medium with B27 in the presence of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) induced their growth and expansion as neurospheres. Addition of all-trans retinoic acid (ATRA) and PPARγ agonist ciglitazone or 15-Deoxy-Δ12,14-Prostaglandin J2 (15d-PGJ2) resulted in a dose-dependent inhibition of cell viability and proliferation of NSCs in culture. Interestingly, NSCs cultured with PPARγ agonists, but not ATRA, showed significant increase in oligodendrocyte precursor-specific O4 and NG2 reactivity with a reduction in NSC marker nestin, in 3–7 days. In vitro treatment with PPARγ agonists and ATRA also induced modest increase in the expression of neuronal β-III tubulin and astrocyte-specific GFAP in NSCs in 3–7 days. Further analyses showed that PPARγ agonists and ATRA induced significant alterations in the expression of many stemness and differentiation genes associated with neuro-glial differentiation in NSCs. These findings highlight the influence of PPARγ agonists in promoting neuro-glial differentiation of NSCs and its significance in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Saravanan Kanakasabai
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
| | - Ecaterina Pestereva
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
| | - Wanida Chearwae
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
| | - Sushil K. Gupta
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
| | - Saif Ansari
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
| | - John J. Bright
- Neuroscience Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, Indiana, United States of America
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
31
|
Chiang MC, Cheng YC, Lin KH, Yen CH. PPARγ regulates the mitochondrial dysfunction in human neural stem cells with tumor necrosis factor alpha. Neuroscience 2012; 229:118-29. [PMID: 23153990 DOI: 10.1016/j.neuroscience.2012.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/19/2012] [Accepted: 11/05/2012] [Indexed: 12/13/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a family of ligand-activated transcription factors, and its ligands are known to control many physiological and pathological conditions. The hypothesis of our study was that the PPARγ agonist (rosiglitazone) could mediate tumor necrosis factor alpha (TNFα) related to the regulation of human neural stem cells (hNSCs), by which TNFα possibly fulfills important roles in neuronal impairment. The results show that PPARγ mediates the cell viability of hNSCs via the downregulation of the activity of caspase 3, indicating that this rescue effect of PPARγ could improve the reduced levels of two mitochondrial regulators, adenosine monophosphate-activated protein kinase (AMPK) and Sirtuin 1 (SIRT1) in the hNSCs with TNFα. The stimulation of mitochondrial function by PPARγ was associated with activation of the PPAR coactivator1 alpha (PGC1α) pathway by up-regulation of oxidative defense and mitochondrial systems. The above protective effects appeared to be exerted by a direct activation of the rosiglitazone, because it protected hNSCs from TNFα-evoked oxidative stress and mitochondrial deficiency. Here we show that the rosiglitazone protects hNSCs against Aβ-induced apoptosis and promotes cell survival. These findings extend our understanding of the central role of PPARγ in TNFα-related neuronal impairment, which probably increases risks of neurodegenerative diseases. The anti-inflammatory effects of PPARγ in the hNSCs with TNFα, and the involved mechanisms were also characterized.
Collapse
Affiliation(s)
- M-C Chiang
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | | | | | | |
Collapse
|
32
|
Babaei R, Javadi-Paydar M, Sharifian M, Mahdavian S, Almasi-Nasrabadi M, Norouzi A, Dehpour AR. Involvement of nitric oxide in pioglitazone memory improvement in morphine-induced memory impaired mice. Pharmacol Biochem Behav 2012; 103:313-21. [PMID: 22944106 DOI: 10.1016/j.pbb.2012.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 08/13/2012] [Accepted: 08/20/2012] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Pioglitazone, a PPAR-γ agonist, which is clinically used in treating diabetic patients, has been recently reported to have crucial roles in improving cognition and memory performance. Since the mechanisms involved in the neuroprotective effect of pioglitazone are not entirely understood, the current study was designed to investigate the possible interaction of pioglitazone with morphine in memory-impaired mice and the probable role of nitric oxide (NO) in this effect. MATERIALS AND METHODS All the experiments were performed in passive avoidance and Y-maze paradigms. To induce memory impairment, mice were administered morphine (1, 3 and 10mg/kg, s.c.) immediately before the training trial. Pioglitazone (20, 40 and 80mg/kg, p.o.) was gavaged 2h prior to the training trial. Further, an NO synthase inhibitor, L-NAME (10mg/kg, i.p.), or an inducible NO synthase inhibitor, aminoguanidine (100mg/kg, i.p.) was administered 30 min before the training trial to determine the possible involvement of NO in the restorative effect of pioglitazone. RESULTS 1) Morphine dose dependently impaired the acquisition of spatial memory and passive avoidance task. 2) Treatment with pioglitazone significantly improved the memory performance in morphine-treated mice in both tests. 3) In the passive avoidance task, L-NAME, but not aminoguanidine, altered the effect of pioglitazone on morphine-induced memory impairment. 4) In Y-maze discrimination, the memory improving effect of pioglitazone was reversed by both NO synthase inhibitors, L-NAME and aminoguanidine. DISCUSSION Our results demonstrate that the pioglitazone improving effect on the morphine-induced impairment of memory acquisition is at least in part through the NO pathway. It is suggested that in short term spatial recognition memory, both inducible and constitutive NO synthases are involved, but in the long term fear memory, only the constitutive NO synthases indicated a prominent role in the anti-amnestic effect of pioglitazone on morphine-induced memory impairment.
Collapse
Affiliation(s)
- Rosa Babaei
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
33
|
Heneka MT, Reyes-Irisarri E, Hüll M, Kummer MP. Impact and Therapeutic Potential of PPARs in Alzheimer's Disease. Curr Neuropharmacol 2012; 9:643-50. [PMID: 22654722 PMCID: PMC3263458 DOI: 10.2174/157015911798376325] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Revised: 02/07/2011] [Accepted: 03/14/2011] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator activated receptors (PPARs) are well studied for their role of peripheral metabolism, but they also may be involved in the pathogenesis of various disorders of the central nervous system (CNS) including multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's and, Parkinson's disease. The observation that PPARs are able to suppress the inflammatory response in peripheral macrophages and in several models of human autoimmune diseases, lead to the idea that PPARs might be beneficial for CNS disorders possessing an inflammatory component. The neuroinflammatory response during the course of Alzheimer's disease (AD) is triggered by the deposition of the β-amyloid peptide in extracellular plaques and ongoing neurodegeneration. Non-steroidal anti-inflammatory drugs (NSAIDs) have been considered to delay the onset and reduce the risk to develop Alzheimer’s disease, while they also directly activate PPARγ. This led to the hypothesis that NSAID protection in AD may be partly mediated by PPARγ. Several lines of evidence have supported this hypothesis, using AD related transgenic cellular and animal models. Stimulation of PPARγ by synthetic agonist (thiazolidinediones) inducing anti-inflammatory, anti-amyloidogenic and insulin sensitizing effects may account for the observed effects. Several clinical trials already revealed promising results using PPARγ agonists, therefore PPARγ represents an attractive therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Michael T Heneka
- University of Bonn, Department of Neurology, Clinical Neurosciences Unit, Bonn, Germany
| | | | | | | |
Collapse
|
34
|
Endothelial cells and astrocytes: a concerto en duo in ischemic pathophysiology. Int J Cell Biol 2012; 2012:176287. [PMID: 22778741 PMCID: PMC3388591 DOI: 10.1155/2012/176287] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 04/30/2012] [Indexed: 11/17/2022] Open
Abstract
The neurovascular/gliovascular unit has recently gained increased attention in cerebral ischemic research, especially regarding the cellular and molecular changes that occur in astrocytes and endothelial cells. In this paper we summarize the recent knowledge of these changes in association with edema formation, interactions with the basal lamina, and blood-brain barrier dysfunctions. We also review the involvement of astrocytes and endothelial cells with recombinant tissue plasminogen activator, which is the only FDA-approved thrombolytic drug after stroke. However, it has a narrow therapeutic time window and serious clinical side effects. Lastly, we provide alternative therapeutic targets for future ischemia drug developments such as peroxisome proliferator- activated receptors and inhibitors of the c-Jun N-terminal kinase pathway. Targeting the neurovascular unit to protect the blood-brain barrier instead of a classical neuron-centric approach in the development of neuroprotective drugs may result in improved clinical outcomes after stroke.
Collapse
|
35
|
He X, Feng L, Meng H, Wang X, Liu S. Rosiglitazone protects dopaminergic neurons against lipopolysaccharide-induced neurotoxicity through inhibition of microglia activation. Int J Neurosci 2012; 122:532-40. [PMID: 22524690 DOI: 10.3109/00207454.2012.686544] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent evidence has suggested that microglia activation plays an important role in the pathogenesis of Parkinson's disease (PD). Activated microglia secrete various proinflammatory cytokines and neurotoxic mediators, which may contribute to the development of PD. Thus, the inhibition of microglia activation may have a therapeutic benefit in the treatment of PD. In the present study, using mesencephalic neuron-microglia mixed culture and microglia-enriched culture, we investigated whether rosiglitazone (RGZ), a member of peroxisome proliferator-activated receptor gamma (PPARγ) agonists, could inhibit microglia activation. Our results showed that RGZ significantly inhibited lipopolysaccharide (LPS)-induced microglia activation and the production of tumor necrosis factor-alpha (TNF-α), nitric oxide (NO), and superoxide. We further investigated the intracellular signaling pathways regulating the production of TNF-α and NO in LPS-activated microglia. The results showed that RGZ inhibited the phosphorylation and nuclear translocation of the p65 subunit of NF-κB, and the phosphorylation of p38 mitogen-activated protein kinase (p38MAPK). Taken together, our results suggested that the therapeutic effects of RGZ were partially mediated by modulating microglia activation.
Collapse
Affiliation(s)
- Xiuquan He
- Department of Anatomy, Shandong University School of Medicine, 44 Wenhua Xilu, Jinan, Shandong, PR China
| | | | | | | | | |
Collapse
|
36
|
Zuhayra M, Zhao Y, von Forstner C, Henze E, Gohlke P, Culman J, Lützen U. Activation of cerebral peroxisome proliferator-activated receptors γ (PPARγ) reduces neuronal damage in the substantia nigra after transient focal cerebral ischaemia in the rat. Neuropathol Appl Neurobiol 2012; 37:738-52. [PMID: 21366664 DOI: 10.1111/j.1365-2990.2011.01169.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIM The function of brain (neuronal) peroxisome proliferator-activated receptor(s) γ (PPARγ) in the delayed degeneration and loss of neurones in the substantia nigra (SN) was studied in rats after transient occlusion of the middle cerebral artery (MCAO). METHODS The PPARγ agonist, pioglitazone, or vehicle was infused intracerebroventricularly over a 5-day period before, during and 5 days after MCAO (90 min). The neuronal degeneration in the SN pars reticularis (SNr) and pars compacta (SNc), the analysis of the number of tyrosine hydroxylase-immunoreactive (TH-IR) neurones and the expression of the PPARγ in these neurones were studied by immunohistochemistry and immunofluorescence staining. The effects of PPARγ activation on excitotoxic and oxidative neuronal damage induced by glutamate and 6-hydroxydopamine were investigated in primary cortical neurones expressing PPARγ. RESULTS Pioglitazone reduced the total and striatal infarct size, neuronal degeneration in both parts of the ipsilateral SN, the loss of TH-IR neurones in the SNc and increased the number of PPARγ-positive TH-IR neurones. Pioglitazone protected primary cortical neurones against oxidative and excitotoxic damage, prevented the loss of neurites and supported the formation of synaptic networks in neurones exposed to glutamate or 6-hydroxydopamine by a PPARγ-dependent mechanism. CONCLUSIONS Activation of cerebral PPARγ confers neuroprotection after ischaemic stroke by preventing both, neuronal damage within the peri-infarct zone and delayed degeneration of neurones and neuronal death in areas remote from the site of ischaemic injury. Pioglitazone and other PPARγ agonists may be useful therapeutic agents to prevent progression of brain damage after cerebral ischaemia.
Collapse
Affiliation(s)
- M Zuhayra
- Department of Nuclear Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Lim HA, Lee EK, Kim JM, Park MH, Kim DH, Choi YJ, Ha YM, Yoon JH, Choi JS, Yu BP, Chung HY. PPARγ activation by baicalin suppresses NF-κB-mediated inflammation in aged rat kidney. Biogerontology 2011; 13:133-45. [PMID: 22033706 DOI: 10.1007/s10522-011-9361-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 10/11/2011] [Indexed: 12/31/2022]
Abstract
Baicalin, a herb-derived flavonoid compound, has beneficial activities, including the modulation of oxidative stress and inflammation. Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated transcription factor that plays an important role in regulating nuclear factor-κB (NF-κB)-induced age-related inflammation. We investigated the anti-inflammatory action of baicalin, which depends on its ability to activate PPARγ, and subsequently to suppress NF-κB. We examined baicalin-treated kidney tissue from 24-month-old Fischer 344 aged rats (10 or 20 mg/kg/day for 10 days) and baicalin-fed mice (10 mg/kg/day for 3 days) for in vivo investigations, and used endothelial YPEN-1 cells for in vitro studies. In the baicalin-fed aged rats, there was a marked enhancement of both nuclear protein levels and DNA binding activity of PPARγ, and a decreased expression of NF-κB target genes (VCAM-1, IL-1β, and IL-6) compared with non-baicalin-fed aged rats. Furthermore, to confirm the anti-inflammatory action of PPARγ activated by baicalin, we used lipopolysaccharide (LPS)-treated cells and mice. The results showed that baicalin induced PPARγ-selective activation in YPEN-1 cells, and that the effects of baicalin were blocked by the PPARγ receptor antagonist, GW9662. In addition, baicalin treatment prevented RS generation, NF-κB activation and the expression of pro-inflammatory genes, whereas it increased PPARγ expression in LPS-treated cells and mouse kidney. Our data suggest that baicalin-induced PPARγ expression reduced age-related inflammation through blocking pro-inflammatory NF-κB activation. These results indicate that baicalin is a novel PPARγ activator and that this agent may have the potential to minimize inflammation.
Collapse
Affiliation(s)
- Hyun Ae Lim
- Molecular Inflammation Research Center for Aging Intervention, Pusan National University, Gumjung-gu, Busan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sadaghiani MS, Javadi-Paydar M, Gharedaghi MH, Fard YY, Dehpour AR. Antidepressant-like effect of pioglitazone in the forced swimming test in mice: The role of PPAR-gamma receptor and nitric oxide pathway. Behav Brain Res 2011; 224:336-43. [DOI: 10.1016/j.bbr.2011.06.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Revised: 06/07/2011] [Accepted: 06/11/2011] [Indexed: 10/18/2022]
|
39
|
Li WL, Liang X, Wang X, Zhang XD, Liu R, Zhang W, Chen HL, Qin XJ, Bai H, Hai CX. Protective effect of the peroxisome proliferator-activated receptor (PPAR)-γ, ligand rosiglitazone on tert-butyl hydroperoxide-induced QZG cell injury. ACTA ACUST UNITED AC 2011; 63:527-33. [DOI: 10.1016/j.etp.2010.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 04/08/2010] [Accepted: 04/12/2010] [Indexed: 01/29/2023]
|
40
|
Peroxisome Proliferator-Activated Receptor-gamma in Amyotrophic Lateral Sclerosis and Huntington's Disease. PPAR Res 2011; 2008:418765. [PMID: 18464922 PMCID: PMC2366134 DOI: 10.1155/2008/418765] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 02/19/2008] [Indexed: 01/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating and one of the most common adult-onset neurodegenerative diseases with the prevalence of about 5 per 100 000 individuals. It results in the progressive loss of upper and lower motor neurons and leads to gradual muscle weakening ultimately causing paralysis and death. ALS has an obscure cause and currently no effective treatment exists. In this review, a potentially important pathway is described that can be activated by peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists and has the ability to block the neuropathological damage caused by inflammation in ALS and possibly in other neudegenerative diseases like Huntington's disease (HD). Neuroinflammation is a common pathological feature in neurodegenerative diseases. Therefore, PPAR-gamma agonists are thought to be neuroprotective in ALS and HD. We and others have tested the neuroprotective effect of pioglitazone (Actos), a PPAR-gamma agonist, in G93A SOD1 transgenic mouse model of ALS and found significant increase in survival of G93A SOD1 mice. These findings suggest that PPAR-gamma may be an important regulator of neuroinflammation and possibly a new target for the development of therapeutic strategies for ALS. The involvement of PPAR-gamma in HD is currently under investigation, one study finds that the treatment with rosiglitazone had no protection in R6/2 transgenic mouse model of HD. PPAR-gamma coactivator-1alpha (PGC-1alpha) is a transcriptional coactivator that works together with combination of other transcription factors like PPAR-gamma in the regulation of mitochondrial biogenesis. Therefore, PPAR-gamma is a possible target for ALS and HD as it functions as transcription factor that interacts with PGC-1alpha. In this review, the role of PPAR-gamma in ALS and HD is discussed based on the current literature and hypotheses.
Collapse
|
41
|
Regulation of Glial Cell Functions by PPAR-gamma Natural and Synthetic Agonists. PPAR Res 2011; 2008:864140. [PMID: 18464925 PMCID: PMC2367430 DOI: 10.1155/2008/864140] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 03/12/2008] [Indexed: 11/18/2022] Open
Abstract
In the recent years, the peroxisome proliferator-activated receptor-γ (PPAR-γ), a well known target for type II diabetes treatment, has received an increasing attention for its therapeutic potential in inflammatory and degenerative brain disorders. PPAR-γ agonists, which include naturally occurring compounds (such as long chain fatty acids and the cyclopentenone prostaglandin 15-deoxy Δ12,14 prostaglandin J2), and synthetic agonists (among which the thiazolidinediones and few nonsteroidal anti-inflammatory drugs) have shown anti-inflammatory and protective effects in several experimental models of Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, multiple sclerosis and stroke, as well as in few clinical studies. The pleiotropic effects of PPAR-γ agonists are likely to be mediated by several mechanisms involving anti-inflammatory activities on peripheral immune cells (macrophages and lymphocytes), as well as direct effects on neural cells including cerebral vascular endothelial cells, neurons, and glia. In the present article, we will review the recent findings supporting a major role for PPAR-γ agonists in controlling neuroinflammation and neurodegeneration through their activities on glial cells, with a particular emphasis on microglial cells as major macrophage population of the brain parenchyma and main actors in brain inflammation.
Collapse
|
42
|
Abstract
Peroxisome proliferator-activated receptors (PPARs) are well studied for their peripheral physiological and pathological impact, but they also play an important role for the pathogenesis of various disorders of the central nervous system (CNS) like multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's, and Parkinson's disease. The observation that PPARs are able to suppress the inflammatory response in peripheral macrophages and in several models of human autoimmune diseases lead to the idea that PPARs might be beneficial for CNS disorders possessing an inflammatory component. The neuroinflammatory response during the course of Alzheimer's disease (AD) is triggered by the neurodegeneration and the deposition of the β-amyloid peptide in extracellular plaques. Nonsteroidal anti-inflammatory drugs (NSAIDs) have been considered to delay the onset and reduce the risk to develop Alzheimer's disease, while they also directly activate PPARγ. This led to the hypothesis that NSAID protection in AD may be partly mediated by PPARγ. Several lines of evidence have supported this hypothesis, using AD-related transgenic cellular and animal models. Stimulation of PPARγ receptors by synthetic agonist (thiazolidinediones) inducing anti-inflammatory, anti-amyloidogenic, and insulin sensitising effects may account for the observed effects. Several clinical trials already revealed promising results using PPAR agonists, therefore PPARs represent an attractive therapeutic target for the treatment of AD.
Collapse
|
43
|
RAR/RXR and PPAR/RXR Signaling in Spinal Cord Injury. PPAR Res 2011; 2007:29275. [PMID: 18060014 PMCID: PMC1950239 DOI: 10.1155/2007/29275] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2006] [Accepted: 02/28/2007] [Indexed: 12/24/2022] Open
Abstract
The retinoid
acid receptors (RAR) and peroxisome proliferator-activated receptors (PPAR)
have been implicated in the regulation of inflammatory reactions. Both receptor families contain ligand-activated transcription factors which form heterodimers with retinoid X receptors (RXR). We review data that imply RAR/RXR and PPAR/RXR pathways in physiological reactions after spinal cord injury. Experiments show how RAR signaling may improve axonal regeneration and modulate reactions of glia cells. While anti-inflammatory properties of PPAR are well documented in the periphery, their possible roles in the central nervous system have only recently become evident. Due to its anti-inflammatory function this transcription factor family promises to be a useful target after spinal cord or brain lesions.
Collapse
|
44
|
Microglia and Astrocyte Activation by Toll-Like Receptor Ligands: Modulation by PPAR-gamma Agonists. PPAR Res 2011; 2008:453120. [PMID: 18584038 PMCID: PMC2435222 DOI: 10.1155/2008/453120] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 05/05/2008] [Indexed: 12/12/2022] Open
Abstract
Microglia and astrocytes express numerous members of the Toll-like receptor (TLR) family that are pivotal for recognizing conserved microbial motifs expressed by a wide array of pathogens. Despite the critical role for TLRs in pathogen recognition, when dysregulated these pathways can also exacerbate CNS tissue destruction. Therefore, a critical balance must be achieved to elicit sufficient immunity to combat CNS infectious insults and downregulate these responses to avoid pathological tissue damage. We performed a comprehensive survey on the efficacy of various PPAR-γ agonists to modulate proinflammatory mediator release from primary microglia and astrocytes in response to numerous TLR ligands relevant to CNS infectious diseases. The results demonstrated differential abilities of select PPAR-γ agonists to modulate glial activation. For example, 15d-PGJ2 and pioglitazone were both effective at reducing IL-12 p40 release by TLR ligand-activated glia, whereas CXCL2 expression was either augmented or inhibited by 15d-PGJ2, effects that were dependent on the TLR ligand examined. Pioglitazone and troglitazone demonstrated opposing actions on microglial CCL2 production that were TLR ligand-dependent. Collectively, this information may be exploited to modulate the host immune response during CNS infections to maximize host immunity while minimizing inappropriate bystander tissue damage that is often characteristic of such diseases.
Collapse
|
45
|
The PPAR-gamma Agonist 15-Deoxy-Delta-Prostaglandin J(2) Attenuates Microglial Production of IL-12 Family Cytokines: Potential Relevance to Alzheimer's Disease. PPAR Res 2011; 2008:349185. [PMID: 18615183 PMCID: PMC2442897 DOI: 10.1155/2008/349185] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2008] [Accepted: 04/22/2008] [Indexed: 01/01/2023] Open
Abstract
Accumulation of amyloid-beta peptide (Abeta) appears to contribute to the pathogenesis of Alzheimer's disease (AD). Therapeutic hope for the prevention or removal of Abeta deposits has been placed in strategies involving immunization against the Abeta peptide. Initial Abeta immunization studies in animal models of AD showed great promise. However, when this strategy was attempted in human subjects with AD, an unacceptable degree of meningoencephalitis occurred. It is generally believed that this adverse outcome resulted from a T-cell response to Abeta. Specifically, CD4(+) Th1 and Th17 cells may contribute to severe CNS inflammation and limit the utility of Abeta immunization in the treatment of AD. Interleukin (IL)-12 and IL-23 play critical roles in the development of Th1 and Th17 cells, respectively. In the present study, Abeta(1-42) synergistically elevated the expression of IL-12 and IL-23 triggered by inflammatory activation of microglia, and the peroxisome proliferator-activated receptor (PPAR)-gamma agonist 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) effectively blocked the elevation of these proinflammatory cytokines. Furthermore, 15d-PGJ(2) suppressed the Abeta-related synergistic induction of CD14, MyD88, and Toll-like receptor 2, molecules that play critical roles in neuroinflammatory conditions. Collectively, these studies suggest that PPAR-gamma agonists may be effective in modulating the development of AD.
Collapse
|
46
|
Morales-Garcia JA, Luna-Medina R, Alfaro-Cervello C, Cortes-Canteli M, Santos A, Garcia-Verdugo JM, Perez-Castillo A. Peroxisome proliferator-activated receptor γ ligands regulate neural stem cell proliferation and differentiation in vitro and in vivo. Glia 2011; 59:293-307. [PMID: 21125653 DOI: 10.1002/glia.21101] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a family of ligand-activated nuclear receptors and its ligands are known to control many physiological and pathological situations. Its role in the central nervous system has been under intense analysis during the last years. Here we show a novel function for PPARγ in controlling stem cell expansion in the adult mammalian brain. Adult rats treated with pioglitazone, a specific ligand of PPARγ, had elevated numbers of proliferating progenitor cells in the subventricular zone and the rostral migratory stream. Electron microscopy analysis also showed important changes in the subventricular zone ultrastructure of pioglitazone-treated animals including an increased number of migratory cell chains. These results were further confirmed in vitro. Neurosphere assays revealed significant increases in the number of neurosphere forming cells from pioglitazone- and rosiglitazone (two specific ligands of PPARγ receptor)-treated cultures that exhibited enhanced capacity for cell migration and differentiation. The effects of pioglitazone were blocked by the PPARγ receptor antagonists GW9662 and T0070907, suggesting that its effects are mediated by a mechanism dependent on PPARγ activation. These results indicate for the first time that activation of PPARγ receptor directly regulates proliferation, differentiation, and migration of neural stem cells in vivo.
Collapse
Affiliation(s)
- Jose A Morales-Garcia
- Instituto de Investigaciones Biomédicas, CSIC-UAM, Arturo Duperier 4, 28029-Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Liu Y, Wu J, Yan C, Liu FR. Correlation of PPARγ and NF-κB expression with arsenic-induced hepatic fibrosis in rats. Shijie Huaren Xiaohua Zazhi 2010; 18:3848-3856. [DOI: 10.11569/wcjd.v18.i36.3848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the significance of peroxisome proliferator activated receptor γ (PPARγ) and nuclear transcription factor kappa B (NF-κB) expression in arsenic-induced hepatic fibrosis in rats.
METHODS: One hundred and ten Sprague-Dawley rats were randomly divided into control group (tap water), model group (100 mg/L NaASO2) and spontaneous recovery group (100 mg/L NaASO2 + tap water). Ten rats of each group were sacrificed on days 30, 60, 90 and 120. Hepatic histological changes and liver fibrosis were evaluated by hematoxylin and eosin staining and Masson staining. The mRNA and protein expression of PPARγ and NF-κB was detected by real-time PCR and Western blot, respectively.
RESULTS: (1) Pathology: With the prolongation of arsenic exposure, hepatic injury became more serious. Compared with the model group, liver injury was significantly relieved on day 30 after one-month arsenic-exposure in the spontaneous recovery, but showed no significant changes on day 30 after two- or three-month arsenic-exposure. (2) mRNA: Compared with the control group, the expression levels of PPARγ mRNA were significantly reduced at different time points in the model group (174.99 ± 41.48 vs 218.40 ± 47.85, 114.55 ± 21.30 vs 218.40 ± 47.85, 64.67 ± 9.83 vs 218.40 ± 47.85, 19.20 ± 16.10 vs218.40 ± 47.85, all P < 0.05). The expression level of PPARγ mRNA on day 30 after one-month arsenic-exposure in the spontaneous recovery group was significantly higher than that in the corresponding model group (215.97 ± 45.96 vs 174.99 ± 41.48, P < 0.05). Compared with the control group, the levels of NF-κB mRNA were significantly elevated at different time points in the model group (65.58 ± 13.17 vs 30.84 ± 15.24, 90.23 ± 15.68 vs 30.84 ± 15.24, 117.95 ± 18.19 vs 30.84 ± 15.24, 172.86 ± 32.92 vs 30.84 ± 15.24, all P < 0.05). The expression of NF-κB mRNA on day 30 after one-month arsenic-exposure in the spontaneous recovery group was significantly higher than that in the corresponding model group (40.45 ± 19.56 vs 65.58 ± 13.17, P < 0.05). (3) Protein: Compared with the control group, the level of PPARγ protein was reduced in the model group. Compared with the control group, the level of NF-κB protein was elevated in the model group. (4) Correlation: There is a negative correlation between PPARγ and NF-κB mRNA expression (r = 0.847, P < 0.05) as well as between PPARγ and NF-κB protein expression (r = 0.529, P < 0.05).
CONCLUSION: Liver tissue injury is aggravated gradually with the prolongation of arsenic exposure. Earlier avoidance of arsenic exposure is associated with a quicker recovery from liver injury. In arsenic-induced hepatic fibrosis, PPARγ mRNA and protein expression is reduced, while that of NF-κB is elevated. There is a negative correlation between PPARγ and NF-κB expression in arsenic-induced hepatic fibrosis.
Collapse
|
48
|
Antonelli A, Ferrari SM, Frascerra S, Pupilli C, Mancusi C, Metelli MR, Orlando C, Ferrannini E, Fallahi P. CXCL9 and CXCL11 chemokines modulation by peroxisome proliferator-activated receptor-alpha agonists secretion in Graves' and normal thyrocytes. J Clin Endocrinol Metab 2010; 95:E413-20. [PMID: 20810571 DOI: 10.1210/jc.2010-0923] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT Peroxisome proliferator-activated receptor (PPAR)-α has been shown to exert immunomodulatory effects in autoimmune disorders. However, until now, no data were present in the literature about the effect of PPARα activation on CXCL9 and CXCL11 chemokines in general or on secretion of these chemokines in thyroid cells. OBJECTIVE AND DESIGN The presence of PPARα and PPARγ has been evaluated by real-time-PCR in Graves' disease (GD) and control cells in primary culture. Furthermore, we have tested the role of PPARα and PPARγ activation on CXCL9 and CXCL11 secretion in GD and control cells after stimulation of these chemokines secretion with IFNγ and TNFα. RESULTS This study shows the presence of PPARα and PPARγ in GD and control cells. A potent dose-dependent inhibition by PPARα-agonists was observed on the cytokines-stimulated secretion of CXCL9 and CXCL11 in GD and control cells. The potency of the PPARα agonists used was maximum on the secretion of CXCL9, reaching about 90% of inhibition by fenofibrate and 85% by ciprofibrate. The relative potency of the compounds was different with each chemokine; for example, gemfibrozil exerted a 55% inhibition on CXCL11, whereas it had a weaker activity on CXCL9 (40% inhibition). PPARα agonists were stronger (ANOVA, P<0.001) inhibitors of CXCL9 and CXCL11 secretion in thyrocytes than PPARγ agonists. CONCLUSIONS Our study shows the presence of PPARα in GD and control thyrocytes. PPARα activators are potent inhibitors of the secretion of CXCL9 and CXCL11, suggesting that PPARα may be involved in the modulation of the immune response in the thyroid.
Collapse
Affiliation(s)
- Alessandro Antonelli
- Department of Internal Medicine, University of Pisa School of Medicine, Via Roma, 67, I-56100, Pisa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Régnier A, Vicaut E, Mraovitch S. Aggravation of seizure-associated microvascular injuries by ibuprofen may involve multiple pathways. Epilepsia 2010; 51:2412-22. [DOI: 10.1111/j.1528-1167.2009.02480.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Švajger U, Obermajer N, Jeras M. Novel Findings in Drug-Induced Dendritic Cell Tolerogenicity. Int Rev Immunol 2010; 29:574-607. [DOI: 10.3109/08830185.2010.522280] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|