1
|
Ahn H, Lee H, Choi W, Lee H, Lee KG, Youn I, Hur W, Han S, Song C. Discovery of the therapeutic potential of naltriben against glutamate-induced neurotoxicity. Neurochem Int 2025; 183:105928. [PMID: 39756586 DOI: 10.1016/j.neuint.2025.105928] [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: 10/11/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Glutamate-induced neuronal death is associated with neurodegeneration including cerebral ischemia. Several μ-opioid receptor antagonists exhibit a neuroprotective activity and have been considered as a potential therapeutic option for neurodegenerative disorders. For the first time, our current study unveiled the neuroprotective activity of selective δ-opioid receptor antagonists. A potent, selective δ-opioid receptor antagonist naltriben, also known as a potent TRPM7 agonist, displayed the prominent protective effect against glutamate-induced toxicity through opioid receptor-independent, TRPM7-independent mechanisms in HT22 cells. Naltriben activated Nrf2 pathway, and alleviated glutamate-induced Ca2+ influx, ROS production, and apoptosis. Moreover, intraperitoneal administration of naltriben at 20 mg/kg greatly reduced the infarct volume in the subcortical photothrombotic ischemia mouse model in vivo. The neuroprotective activity of naltriben was enhanced by a longer pretreatment, indicating that like Nrf2 activators, naltriben also requires the cellular priming for its full protective effects. Together, these results suggested naltriben as a potential therapeutic agent in conditions related with glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- Hyomin Ahn
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Life Sciences, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyomin Lee
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wonseok Choi
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyebin Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Kang-Gon Lee
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Inchan Youn
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Wooyoung Hur
- Medicinal Materials Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; HY-KIST Bioconvergence, Hanyang University, 222 Wangsimniro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Sungmin Han
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; KHU-KIST, Department of Converging Science and Technology, Kyung Hee University, 26 Kyungheedaero, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Chiman Song
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Hwarangro 14 Gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
2
|
Chemello C, Facci L, Marcolin E, Ramaschi GE, Barbierato M, Giusti P, Bolego C, Zusso M. Fentanyl enhances immune cell response through TLR4/MD-2 complex. Front Pharmacol 2024; 15:1468644. [PMID: 39444612 PMCID: PMC11496304 DOI: 10.3389/fphar.2024.1468644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Opioids have been shown to induce neuroinflammation and immune cell activation, that might contribute to some of the opioid side effects, such as opioid-induced tolerance and paradoxical hyperalgesia. In this context, TLR4/MD-2 complex has been proposed as an off-target site for opioid action. This study was aimed at investigating the effect of fentanyl on lipopolysaccharide (LPS)-induced TLR4/MD-2 activation in rat primary microglia and human monocyte-derived macrophages (MDM). Materials and Methods The effect of fentanyl was first explored by measuring the expression and release of different proinflammatory mediators in primary rat microglia and human MDM by real-time PCR and ELISA. Then, the involvement of TLR4/MD-2 signaling was investigated studying NF-κB activation in HEK293 cells stably transfected with human TLR4, MD-2, and CD14 genes (HEK-Blue hTLR4 cells) and in human MDM. Results Fentanyl increased mRNA levels, as well as the LPS-induced secretion of proinflammatory mediators in primary microglia and MDM. Two inhibitors of TLR4/MD-2 signaling, namely the oxazoline derivative of N-palmitoylethanolamine (PEA-OXA) and CLI-095, blocked the production and release of proinflammatory cytokines by microglia stimulated with LPS and fentanyl, suggesting that TLR4/MD-2 could be the target of the proinflammatory activity of fentanyl. Finally, we showed that fentanyl in combination with LPS activated NF-κB signaling in human MDM and in HEK-Blue hTLR4 cells and this effect was blocked by inhibitors of TLR4/MD-2 complex. Discussion These results provide new insight into the mechanism of the proinflammatory activity of fentanyl, which involves the activation of TLR4/MD-2 signaling. Our findings might facilitate the development of novel inhibitors of TLR4/MD-2 signaling to combine with opioid-based analgesics for effective and safe pain management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
3
|
Mitchell CL, Kurouski D. Novel strategies in Parkinson's disease treatment: a review. Front Mol Neurosci 2024; 17:1431079. [PMID: 39183754 PMCID: PMC11341544 DOI: 10.3389/fnmol.2024.1431079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
An unprecedented extension of life expectancy observed during the past century drastically increased the number of patients diagnosed with Parkinson's diseases (PD) worldwide. Estimated costs of PD alone reached $52 billion per year, making effective neuroprotective treatments an urgent and unmet need. Current treatments of both AD and PD focus on mitigating the symptoms associated with these pathologies and are not neuroprotective. In this review, we discuss the most advanced therapeutic strategies that can be used to treat PD. We also critically review the shift of the therapeutic paradigm from a small molecule-based inhibition of protein aggregation to the utilization of natural degradation pathways and immune cells that are capable of degrading toxic amyloid deposits in the brain of PD patients.
Collapse
Affiliation(s)
- Charles L. Mitchell
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Dmitry Kurouski
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
4
|
Beygi M, Shayegh J, Esmaeili Gouvarchin Ghaleh H. Caffeine and naloxone treated mesenchymal stem cells improve symptoms and reduce inflammation in a mouse model of ulcerative colitis. Transpl Immunol 2024; 82:101986. [PMID: 38184213 DOI: 10.1016/j.trim.2024.101986] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
BACKGROUND AND OBJECTIVE Ulcerative colitis (UC) causes ulcers in the colon and rectum, leading to abdominal pain, diarrhea, and rectal bleeding, and if left untreated, can lead to serious complications. The therapeutic effects of mesenchymal stem cells (MSCs) on experimental models of UC have been proven. Since the microenvironment around these cells is crucial in maintaining cell proliferation, differentiation, metabolism, and overall function, this study aims to evaluation the role of caffeine and naloxone as a new microenvironment for MSCs in reducing inflammation and improving symptoms in an experimental model of UC. MATERIAL AND METHOD A group of 40 outbred NMRI mice were studied and divided randomly into four equal groups (N = 10 each group). UC was induced in all groups using acetic acid. The first group (control) was treated with phosphate buffer saline (PBS), the second group with MSCs-Caffeine, the third with MSCs-Naloxone, and the fourth with Mesalazine. The disease activity index (DAI), tissue damage, myeloperoxidase (MPO) activity, nitric oxide (NO) levels, and the production of IL-1, IL-6, and TNF-α cytokines were evaluated. RESULT Our research demonstrated that all treatments were effective in improving the symptoms and reducing inflammatory markers in mice with colitis. Among the two MSCs treatments, the MSCs-Caffeine was found to be the most potent in reducing the levels of NO, IL-1, IL-6, tissue damage (P < 0.001) and as well as TNF-α (P < 0.0001) in compared to the control group. CONCLUSION MSCs treated with caffeine and naloxone can enhance the immunoregulatory potential of these. As a result, treated MSCs can lead to improved clinical signs and reduced inflammatory parameters in mice with UC, making this approach a useful way for controlling and treating the disease. However, additional research is needed to access the mechanism behind the stronger immune system regulatory effects of treated MSCs in UC treatment.
Collapse
Affiliation(s)
- Milad Beygi
- Department of Veterinary Medicine, Shabestar Branch, Islamic Azad University, Shabestar, Iran
| | - Jalal Shayegh
- Department of Veterinary Medicine, Shabestar Branch, Islamic Azad University, Shabestar, Iran
| | | |
Collapse
|
5
|
Dong S, Chen C, Di C, Wang S, Dong Q, Lin W, Liu D. The Association between NADPH Oxidase 2 (NOX2) and Drug Resistance in Cancer. Curr Cancer Drug Targets 2024; 24:1195-1212. [PMID: 38362697 DOI: 10.2174/0115680096277328240110062433] [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: 08/16/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 02/17/2024]
Abstract
NADPH oxidase, as a major source of intracellular reactive oxygen species (ROS), assumes an important role in the immune response and oxidative stress response of the body. NADPH oxidase 2 (NOX2) is the first and most representative member of the NADPH oxidase family, and its effects on the development of tumor cells are gaining more and more attention. Our previous study suggested that NCF4 polymorphism in p40phox, a key subunit of NOX2, affected the outcome of diffuse large B-cell lymphoma patients treated with rituximab. It hypothesized that NOX2-mediated ROS could enhance the cytotoxic effects of some anti-tumor drugs in favor of patients with tumors. Several reviews have summarized the role of NOX2 and its congeners-mediated ROS in anti-tumor therapy, but few studies focused on the relationship between the expression of NOX2 and anti-tumor drug resistance. In this article, we systematically introduced the NOX family, represented by NOX2, and a classification of the latest inhibitors and agonists of NOX2. It will help researchers to have a more rational and objective understanding of the dual role of NOX2 in tumor drug resistance and is expected to provide new ideas for oncology treatment and overcoming drug resistance in cancer.
Collapse
Affiliation(s)
- Shiqi Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Chao Chen
- Department of laboratory, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Chang Di
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Shufan Wang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Quan Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Wenxin Lin
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Duo Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| |
Collapse
|
6
|
Cipriano A, Viviano M, Feoli A, Milite C, Sarno G, Castellano S, Sbardella G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J Med Chem 2023; 66:11632-11655. [PMID: 37650225 PMCID: PMC10510401 DOI: 10.1021/acs.jmedchem.3c00770] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 09/01/2023]
Abstract
NADPH oxidases (NOXs) form a family of electron-transporting membrane enzymes whose main function is reactive oxygen species (ROS) generation. Strong evidence suggests that ROS produced by NOX enzymes are major contributors to oxidative damage under pathologic conditions. Therefore, blocking the undesirable actions of these enzymes is a therapeutic strategy for treating various pathological disorders, such as cardiovascular diseases, inflammation, and cancer. To date, identification of selective NOX inhibitors is quite challenging, precluding a pharmacologic demonstration of NOX as therapeutic targets in vivo. The aim of this Perspective is to furnish an updated outlook about the small-molecule NOX inhibitors described over the last two decades. Structures, activities, and in vitro/in vivo specificity are discussed, as well as the main biological assays used.
Collapse
Affiliation(s)
- Alessandra Cipriano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Monica Viviano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Alessandra Feoli
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Ciro Milite
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Giuliana Sarno
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Sabrina Castellano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Gianluca Sbardella
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| |
Collapse
|
7
|
NOX2 oxidase inhibitor GSK2795039 possess antiviral activity against H1N1 influenza A virus in vitro and vivo. Microb Pathog 2023; 174:105942. [PMID: 36502994 DOI: 10.1016/j.micpath.2022.105942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
The continuous zoonotic circulation and reassortment potential of influenza A viruses (IAV) in nature represents an enormous public health threat to humans. Beside vaccination antivirals are needed to efficiently control spreading of the disease. The previous research has shown that NOX2 involved in IAV replication, but the detailed mechanism has not been reported. In the present study we investigated the roles of NOX2 in host inflammatory response and IAV replication using a novel inhibitor GSK2795039. The drug significantly reduced H1N1 virus induced NOX2 activity and ROS release in human lung epithelial cells. The results of time course experiments suggested that GSK2795039 inhibited an early post-entry step of viral infection. Concomitantly, there was a decreased expression of pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interferon (IFN)-β and interleukin (IL)-6) in NOX2 suppressed cells. In vivo, compared with control groups, suppression of NOX2 improved the survival rate of mice infected with H1N1 virus (42.9% in GSK2795039 treated mice versus >0% of control mice) and viral burden also decreased in the GSK2795039 treated group. Thus, our data demonstrated a critical role for NOX2 in the establishment of H1N1 infection and subsequent inflammatory reactions, which suggest that GSK2795039 may be a potential therapeutic drug for IAV infection.
Collapse
|
8
|
Chen SH, Han S, Hu CF, Zhou R, Gao Y, Tu D, Gao H, Feng J, Wang Y, Lu RB, Hong JS. Activation of the MAC1-ERK1/2-NOX2 Pathway Is Required for LPS-Induced Sustaining Reactive Microgliosis, Chronic Neuroinflammation and Neurodegeneration. Antioxidants (Basel) 2022; 11:1202. [PMID: 35740099 PMCID: PMC9220294 DOI: 10.3390/antiox11061202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/24/2022] Open
Abstract
Recent studies suggest that improper resolution of acute neuroinflammation may lead to long-lasting low-grade chronic neuroinflammation and drive progressive neurodegeneration. However, the molecular mechanism underlying the transition from acute to chronic neuroinflammation remains unclear. The main purpose of this study was to search for potential pathways mediating LPS-elicited chronic neuroinflammation and resultant neurodegeneration. Using microglia cultures prepared from C57BL/6J, MAC1-deficient, and MyD88-deficient mice, the initial study showed that activation of TLR-4 is not sufficient for maintaining chronic neuroinflammation despite its essential role in LPS-initiated acute neuroinflammation. Opposite to TLR-4, our studies showed significantly reduced intensity of chronic neuroinflammation, oxidative stress, and progressive loss of nigral dopaminergic neurons in MAC1-deficient neuron/glial cultures or mice stimulated with LPS. Mechanistic studies revealed the essential role ERK1/2 activation in chronic neuroinflammation-elicited neurodegeneration, which was demonstrated by using an ERK1/2 inhibitor in neuron-glial cultures. Taken together, we propose a key role of the MAC1-NOX2-ERK1/2 signaling pathway in the initiation and maintenance of low-grade chronic neuroinflammation. Continuing ERK1/2 phosphorylation and NOX2 activation form a vicious feedforward cycle in microglia to maintain the low-grade neuroinflammation and drive neurodegeneration.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
| | - Shuangyu Han
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Chih-Fen Hu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan
| | - Ran Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Yun Gao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Dezhen Tu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Huiming Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Yubao Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Institute of Infectious Diseases, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ru-Band Lu
- Institute of Behavioral Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
| |
Collapse
|
9
|
Reducing neuroinflammation via therapeutic compounds and lifestyle to prevent or delay progression of Parkinson's disease. Ageing Res Rev 2022; 78:101618. [PMID: 35395416 DOI: 10.1016/j.arr.2022.101618] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/08/2022] [Accepted: 04/01/2022] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is the second most common age-associated neurodegenerative disorder and is characterised by progressive loss of dopamine neurons in the substantia nigra. Peripheral immune cell infiltration and activation of microglia and astrocytes are observed in PD, a process called neuroinflammation. Neuroinflammation is a fundamental response to protect the brain but, when chronic, it triggers neuronal damage. In the last decade, central and peripheral inflammation were suggested to occur at the prodromal stage of PD, sustained throughout disease progression, and may play a significant role in the pathology. Understanding the pathological mechanisms of PD has been a high priority in research, primarily to find effective treatments once symptoms are present. Evidence indicates that early life exposure to neuroinflammation as a consequence of life events, environmental or behaviour factors such as exposure to infections, pollution or a high fat diet increase the risk of developing PD. Many studies show healthy habits and products that decrease neuroinflammation also reduce the risk of PD. Here, we aim to stimulate discussion about the role of neuroinflammation in PD onset and progression. We highlight that reducing neuroinflammation throughout the lifespan is critical for preventing idiopathic PD, and present epidemiological studies that detail risk and protective factors. It is possible that introducing lifestyle changes that reduce neuroinflammation at the time of PD diagnosis may slow symptom progression. Finally, we discuss compounds and therapeutics to treat the neuroinflammation associated with PD.
Collapse
|
10
|
Peyravian N, Sun E, Dikici E, Deo S, Daunert S, Toborek M. Opioid Antagonist Nanodrugs Successfully Attenuate the Severity of Ischemic Stroke. Mol Pharm 2022; 19:2254-2267. [PMID: 35506882 PMCID: PMC9257743 DOI: 10.1021/acs.molpharmaceut.2c00079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The United States
is in the midst of an opioid epidemic that is
linked to a number of serious health issues, including an increase
in cerebrovascular events, namely, stroke. Chronic prescription opioid
use exacerbates the risk and severity of ischemic stroke, contributing
to stroke as the fifth overall cause of death in the United States
and costing the US health care system over $30 billion annually. Pathologically,
opioids challenge the integrity of the blood–brain barrier
(BBB), resulting in a dysregulation of tight junction (TJ) proteins
that are crucial in maintaining barrier homeostasis. Despite this,
treatment options for ischemic stroke are limited, and there are no
pharmacological options to attenuate TJ damage, including in incidents
that are linked to opioid use. Herein, we have generated carrier-free,
pure “nanodrugs” or nanoparticles of naloxone and naltrexone
with enhanced therapeutic properties compared to the original (parent)
drugs. The generated nanoformulations of both opioid antagonists exhibited
successful attenuation of morphine- or oxycodone-induced alterations
of TJ protein expression and reduced oxidative stress to a greater
extent than the parent drugs (non-nano). As a proof of concept, we
then proceeded to evaluate the therapeutic effectiveness of the generated
nanodrugs in an ischemic stroke model of mice exposed to morphine
or oxycodone. Our results demonstrate that the opioid antagonist nanoformulations
reduced stroke severity in mice. Overall, this research implements
advances in nanotechnology-based repurposing of FDA-approved therapeutics,
and the obtained results also suggest underlying pharmacological mechanisms
of opioid antagonists, further supporting these opioid antagonists
and their respective nanoformulations as potential therapeutic agents
for ischemic stroke.
Collapse
Affiliation(s)
- Nadia Peyravian
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States.,Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, Miami, Florida 33136, United States
| | - Enze Sun
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States.,Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, Miami, Florida 33136, United States
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States.,Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, Miami, Florida 33136, United States.,University of Miami Clinical and Translational Science Institute, Miami, Florida 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States.,Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, Miami, Florida 33136, United States.,University of Miami Clinical and Translational Science Institute, Miami, Florida 33136, United States
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States.,Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, Miami, Florida 33136, United States
| |
Collapse
|
11
|
Cheng WJ, Chiang CC, Lin CY, Chen YL, Leu YL, Sie JY, Chen WL, Hsu CY, Kuo JJ, Hwang TL. Astragalus mongholicus Bunge Water Extract Exhibits Anti-inflammatory Effects in Human Neutrophils and Alleviates Imiquimod-Induced Psoriasis-Like Skin Inflammation in Mice. Front Pharmacol 2021; 12:762829. [PMID: 34955833 PMCID: PMC8707293 DOI: 10.3389/fphar.2021.762829] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Neutrophils are the primary immune cells in innate immunity, which are related to various inflammatory diseases. Astragalus mongholicus Bunge is a Chinese medicinal herb used to treat various oxidative stress-related inflammatory diseases. However, there are limited studies that elucidate the effects of Astragalus mongholicus Bunge in human neutrophils. In this study, we used isolated human neutrophils activated by various stimulants to investigate the anti-inflammatory effects of Astragalus mongholicus Bunge water extract (AWE). Cell-free assays were used to examine free radicals scavenging capabilities on superoxide anion, reactive oxygen species (ROS), and nitrogen-centered radicals. Imiquimod (IMQ) induced psoriasis-like skin inflammation mouse model was used for investigating anti-psoriatic effects. We found that AWE inhibited superoxide anion production, ROS generation, and elastase release in human neutrophils, which exhibiting a direct anti-neutrophil effect. Moreover, AWE exerted a ROS scavenging ability in the 2,2’-Azobis (2-amidinopropane) dihydrochloride assay, but not superoxide anion in the xanthine/xanthine oxidase assay, suggesting that AWE exhibited anti-oxidation and anti-inflammatory capabilities by both scavenging ROS and by directly inhibiting neutrophil activation. AWE also reduced CD11b expression and adhesion to endothelial cells in activated human neutrophils. Meanwhile, in mice with psoriasis-like skin inflammation, administration of topical AWE reduced both the affected area and the severity index score. It inhibited neutrophil infiltration, myeloperoxidase release, ROS-induced damage, and skin proliferation. In summary, AWE exhibited direct anti-inflammatory effects by inhibiting neutrophil activation and anti-psoriatic effects in mice with IMQ-induced psoriasis-like skin inflammation. Therefore, AWE could potentially be a pharmaceutical Chinese herbal medicine to inhibit neutrophilic inflammation for anti-psoriasis.
Collapse
Affiliation(s)
- Wei-Jen Cheng
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Chao Chiang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Puxin Fengze Chinese Medicine Clinic, Taoyuan, Taiwan
| | - Cheng-Yu Lin
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Li Chen
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-Yu Sie
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Ling Chen
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Yuan Hsu
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jong-Jen Kuo
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Traditional Chinese Medicine, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
12
|
Wu R, Li JX. Toll-Like Receptor 4 Signaling and Drug Addiction. Front Pharmacol 2020; 11:603445. [PMID: 33424612 PMCID: PMC7793839 DOI: 10.3389/fphar.2020.603445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
The emphasis of neuronal alterations and adaptations have long been the main focus of the studies of the mechanistic underpinnings of drug addiction. Recent studies have begun to appreciate the role of innate immune system, especially toll-like receptor 4 (TLR4) signaling in drug reward-associated behaviors and physiology. Drugs like opioids, alcohol and psychostimulants activate TLR4 signaling and subsequently induce proinflammatory responses, which in turn contributes to the development of drug addiction. Inhibition of TLR4 or its downstream effectors attenuated the reinforcing effects of opioids, alcohol and psychostimulants, and this effect is also involved in the withdrawal and relapse-like behaviors of different drug classes. However, conflicting results also argue that TLR4-related immune response may play a minimal part in drug addiction. This review discussed the preclinical evidence that whether TLR4 signaling is involved in multiple drug classes action and the possible mechanisms underlying this effect. Moreover, clinical studies which examined the potential efficacy of immune-base pharmacotherapies in treating drug addiction are also discussed.
Collapse
Affiliation(s)
- Ruyan Wu
- School of Medicine, Yangzhou University, Yangzhou, China.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
13
|
Teixeira-Santos L, Albino-Teixeira A, Pinho D. Neuroinflammation, oxidative stress and their interplay in neuropathic pain: Focus on specialized pro-resolving mediators and NADPH oxidase inhibitors as potential therapeutic strategies. Pharmacol Res 2020; 162:105280. [PMID: 33161139 DOI: 10.1016/j.phrs.2020.105280] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a chronic condition that results from a lesion or disease of the nervous system, greatly impacting patients' quality of life. Current pharmacotherapy options deliver inadequate and/or insufficient responses and thus a significant unmet clinical need remains for alternative treatments in NP. Neuroinflammation, oxidative stress and their reciprocal relationship are critically involved in NP pathophysiology. In this context, new pharmacological approaches, aiming at enhancing the resolution phase of inflammation and/or restoring redox balance by targeting specific reactive oxygen species (ROS) sources, are emerging as potential therapeutic strategies for NP, with improved efficacy and safety profiles. Several reports have demonstrated that administration of exogenous specialized pro-resolving mediators (SPMs) ameliorates NP pathophysiology. Likewise, deletion or inhibition of the ROS-generating enzyme NADPH oxidase (NOX), particularly its isoforms 2 and 4, results in beneficial effects in NP models. Notably, SPMs also modulate oxidative stress and NOX also regulates neuroinflammation. By targeting neuroinflammatory and oxidative pathways, both SPMs analogues and isoform-specific NOX inhibitors are promising therapeutic strategies for NP.
Collapse
Affiliation(s)
- Luísa Teixeira-Santos
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| | - António Albino-Teixeira
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| | - Dora Pinho
- Departamento de Biomedicina - Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Portugal; MedInUP - Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto, Portugal.
| |
Collapse
|
14
|
Abstract
Significance: The oxidative stress, resulting from an imbalance in the production and scavenging of reactive oxygen species (ROS), is known to be involved in the development and progression of several pathologies. The excess of ROS production is often due to an overactivation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) and for this reason these enzymes became promising therapeutic targets. However, even if NOX are now well characterized, the development of new therapies is limited by the lack of highly isoform-specific inhibitors. Recent Advances: In the past decade, several groups and laboratories have screened thousands of molecules to identify new specific inhibitors with low off-target effects. These works have led to the characterization of several new potent NOX inhibitors; however, their specificity varies a lot depending on the molecules. Critical Issues: Here, we are reviewing more than 25 known NOX inhibitors, focusing mainly on the newly identified ones such as APX-115, NOS31, Phox-I1 and 2, GLX7013114, and GSK2795039. To have a better overall view of these molecules, the inhibitors were classified according to their specificity, from pan-NOX inhibitors to highly isoform-specific ones. We are also presenting the use of these compounds both in vitro and in vivo. Future Directions: Several of these new molecules are potent and very specific inhibitors that could be good candidates for the development of new drugs. Even if the results are very promising, most of these compounds were only validated in vitro or in mice models and further investigations will be required before using them as potential therapies.
Collapse
Affiliation(s)
- Mathieu Chocry
- Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS, Marseille, France
| | - Ludovic Leloup
- Aix-Marseille Université, Institut de Neurophysiopathologie (INP), CNRS, Marseille, France
| |
Collapse
|
15
|
Hou L, Zhang L, Hong JS, Zhang D, Zhao J, Wang Q. Nicotinamide Adenine Dinucleotide Phosphate Oxidase and Neurodegenerative Diseases: Mechanisms and Therapy. Antioxid Redox Signal 2020; 33:374-393. [PMID: 31968994 DOI: 10.1089/ars.2019.8014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: The growing incidence of neurodegenerative diseases significantly impacts the individuals who suffer from these disorders and is a major health concern globally. Although the specific mechanisms of neurodegenerative diseases are still far from being acknowledged, it is becoming clear that oxidative stress and neuroinflammation are critical contributing factors to the progression of neurodegeneration. Thus, it is conceivable that the inhibition of oxidative stress and neuroinflammation may represent promising therapeutic targets for the treatment of neurodegenerative diseases. Recent Advances: Recently, the strategy for neurodegenerative disease therapy has shifted from the use of antioxidants and conventional anti-inflammatory targets to upstream mediators due to the failure of most antioxidants and nonsteroidal anti-inflammatory drugs in clinical trials. Nicotinamide adenine dinucleotide phosphate oxidases (NOXs), a family of superoxide-producing enzyme complexes, have been identified as an upstream factor that controls both oxidative stress and neuroinflammation. Genetic inactivation or pharmacological inhibition of NOX enzymes displays potent neuroprotective effects in a broad spectrum of neurodegenerative disease models. Critical Issues: The detailed mechanisms of how NOX enzymes regulate oxidative stress and neuroinflammation still remain unclear. Moreover, the currently available inhibitors of NOX enzymes exhibit nonspecificity, off-target effects, unsuitable pharmacokinetic properties, and even high toxicity, markedly limiting their potential clinical applications. Future Directions: This review provides novel insights into the roles of NOXs in neurodegenerative pharmacology, and indicates the types of NOX enzyme inhibitors that should be identified and developed as candidates for future applications, which might reveal novel neurodegenerative disease therapies based on NOXs.
Collapse
Affiliation(s)
- Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Dan Zhang
- State Key Laboratory of Natural Products and Functions, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Comparison of three analytical methods for superoxide produced by activated immune cells. J Pharmacol Toxicol Methods 2019; 101:106637. [PMID: 31678429 DOI: 10.1016/j.vascn.2019.106637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/29/2019] [Accepted: 09/30/2019] [Indexed: 11/23/2022]
Abstract
Superoxide plays a key role in normal immune function and inflammatory diseases. In order to evaluate normal immune function or screen inhibitors of superoxide production for treating inflammatory diseases, it is very important to detect superoxide with good accuracy, sensitivity, and flexibility. In present study, we investigated three analysis methods of superoxide, colorimetric assay by WST-8, fluorescence assay by dihydroethidium and chemiluminescence assay by lucigenin, compared their precisions, specificities, sensitivities and time curve characteristics in superoxide analysis, and then validate their values in the screening of anti-inflammatory compounds. The results reveal that three analysis methods of superoxide all have good precisions and high specificities but have different sensitivities and time curve characteristics, which suggest their different applications. In addition, they can all be used in the screening of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors and anti-inflammatory compounds.
Collapse
|
17
|
Peyravian N, Dikici E, Deo S, Toborek M, Daunert S. Opioid antagonists as potential therapeutics for ischemic stroke. Prog Neurobiol 2019; 182:101679. [PMID: 31398359 PMCID: PMC6814577 DOI: 10.1016/j.pneurobio.2019.101679] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/31/2019] [Indexed: 01/09/2023]
Abstract
Chronic use of prescription opioids exacerbates risk and severity of ischemic stroke. Annually, 6 million people die from stroke worldwide and there are no neuroprotective or neurorestorative agents to improve stroke outcomes and promote recovery. Prescribed opioids such as morphine have been shown to alter tight junction protein expression, resulting in the disruption of the blood brain barrier (BBB), ultimately leading to stroke pathogenesis. Consequently, protection of the BBB has been proposed as a therapeutic strategy for ischemic stroke. This perspective addresses the deficiency in stroke pharmacological options and examines a novel application and repurposing of FDA-approved opioid antagonists as a prospective neuroprotective therapeutic strategy to minimize BBB damage, reduce stroke severity, and promote neural recovery. Future directions discuss potential drug design and delivery methods to enhance these novel therapeutic targets.
Collapse
Affiliation(s)
- Nadia Peyravian
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA.
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, USA; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami, USA; University of Miami Clinical and Translational Science Institute, USA.
| |
Collapse
|
18
|
TLR-2 mediated cytosolic-Ca 2+ surge activates ER-stress-superoxide-NO signalosome augmenting TNF-α production leading to apoptosis of Mycobacterium smegmatis-infected fish macrophages. Sci Rep 2019; 9:12330. [PMID: 31444398 PMCID: PMC6707155 DOI: 10.1038/s41598-019-48847-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/05/2019] [Indexed: 01/14/2023] Open
Abstract
The implications of TLR-2 mediated alterations in cytosolic-Ca2+((Ca2+)c) levels in M. smegmatis infections is not well known. Using headkidney macrophages (HKM) from Clarias gariepinus, we observed TLR-2 signalling is required in the phagocytosis of M. smegmatis. M. smegmatis induced caspase-dependent HKM apoptosis in MOI, time and growth-phase dependent manner. RNAi and inhibitor studies demonstrated critical role of TLR-2 in eliciting (Ca2+)c-surge and c-Src-PI3K-PLC axis playing an intermediary role in the process. The (Ca2+)c-surge triggered downstream ER-stress and superoxide (O2−) generation. The cross-talk between ER-stress and O2− amplified TNF-α production, which led to HKM apoptosis and bacterial clearance. Release of nitric oxide (NO) was also observed and silencing the NOS2-NO axis enhanced intracellular bacterial survival and attenuated caspase activity. Pre-treatment with diphenyleneidonium chloride inhibited NO production implicating O2−–NO axis imperative in M. smegmatis-induced HKM apoptosis. NO positively impacted CHOP expression and TNF-α production in infected HKM. We conclude that, TLR-2 induced (Ca2+)c-surge and ensuing cross-talk between ER-stress and O2− potentiates HKM pathology by amplifying pro-inflammatory TNF-α production. Moreover, the pro-oxidant environment triggers NO release which prolonged ER-stress and TNF-α production, culminating in HKM apoptosis and bacterial clearance. Together, our study suggests HKM an alternate model to study macrophage-mycobacteria interactions.
Collapse
|
19
|
Hou L, Sun F, Huang R, Sun W, Zhang D, Wang Q. Inhibition of NADPH oxidase by apocynin prevents learning and memory deficits in a mouse Parkinson's disease model. Redox Biol 2019; 22:101134. [PMID: 30798073 PMCID: PMC6389731 DOI: 10.1016/j.redox.2019.101134] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/28/2019] [Accepted: 02/03/2019] [Indexed: 12/29/2022] Open
Abstract
The activation of NADPH oxidase contributes to dopaminergic neurodegeneration and motor deficits in Parkinson's disease (PD). However, whether NADPH oxidase is involved in non-motor symptoms, especially cognitive dysfunction in PD remains unknown. This study is undertaken to characterize the effects of inhibition of NADPH oxidase by a widely used NADPH oxidase inhibitor apocynin on learning and memory deficits in paraquat and maneb-induced mouse PD model. Results showed that mice injected with paraquat and maneb displayed impairments of spatial learning and memory, which was associated with reduced tyrosine hydroxylase expression as well as increased neurodegeneration, synaptic loss, α-synuclein expression and Ser129-phosphorylation in the hippocampus. Interestingly, apocynin treatment significantly ameliorated learning and memory deficits as well as hippocampal neurodegeneration and α-synuclein pathology in mice treated with these two pesticides. Mechanistically, we found that apocynin mitigated paraquat and maneb-induced NADPH oxidase activation and related oxidative stress. Furthermore, reduced microglial activation and M1 polarization were observed in apocynin and paraquat and maneb co-treated mice compared with paraquat and maneb alone group. Finally, apocynin inhibited the activation of signal transducers and activators of transcription 1 (STAT1) and nuclear factor kappa B (NF-κB) pathways, two key regulatory factors for microglial M1 inflammatory responses, in paraquat and maneb-treated mice. Altogether, our findings implied that NADPH oxidase mediates learning and memory deficits in PD, and inhibition of NADPH oxidase by apocynin blocks impairments of learning and memory via the suppression of oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Liyan Hou
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Fuqiang Sun
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Ruixue Huang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Wei Sun
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Dan Zhang
- State Key Laboratory of Natural Products and Functions, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
20
|
Post-stroke Intranasal (+)-Naloxone Delivery Reduces Microglial Activation and Improves Behavioral Recovery from Ischemic Injury. eNeuro 2018; 5:eN-NWR-0395-17. [PMID: 29766045 PMCID: PMC5952324 DOI: 10.1523/eneuro.0395-17.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is the leading cause of disability, and effective therapeutic strategies are needed to promote complete recovery. Neuroinflammation plays a significant role in stroke pathophysiology, and there is limited understanding of how it affects recovery. The aim of this study was to characterize the spatiotemporal expression profile of microglial activation and whether dampening microglial/macrophage activation post-stroke facilitates the recovery. For dampening microglial/macrophage activation, we chose intranasal administration of naloxone, a drug that is already in clinical use for opioid overdose and is known to decrease microglia/macrophage activation. We characterized the temporal progression of microglia/macrophage activation following cortical ischemic injury in rat and found the peak activation in cortex 7 d post-stroke. Unexpectedly, there was a chronic expression of phagocytic cells in the thalamus associated with neuronal loss. (+)-Naloxone, an enantiomer that reduces microglial activation without antagonizing opioid receptors, was administered intranasally starting 1 d post-stroke and continuing for 7 d. (+)-Naloxone treatment decreased microglia/macrophage activation in the striatum and thalamus, promoted behavioral recovery during the 14-d monitoring period, and reduced neuronal death in the lesioned cortex and ipsilateral thalamus. Our results are the first to show that post-stroke intranasal (+)-naloxone administration promotes short-term functional recovery and reduces microglia/macrophage activation. Therefore, (+)-naloxone is a promising drug for the treatment of ischemic stroke, and further studies should be conducted.
Collapse
|
21
|
Chen SH, Sung YF, Oyarzabal EA, Tan YM, Leonard J, Guo M, Li S, Wang Q, Chu CH, Chen SL, Lu RB, Hong JS. Physiological Concentration of Prostaglandin E 2 Exerts Anti-inflammatory Effects by Inhibiting Microglial Production of Superoxide Through a Novel Pathway. Mol Neurobiol 2018; 55:8001-8013. [PMID: 29492849 DOI: 10.1007/s12035-018-0965-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/16/2018] [Indexed: 01/21/2023]
Abstract
This study investigated the physiological regulation of brain immune homeostasis in rat primary neuron-glial cultures by sub-nanomolar concentrations of prostaglandin E2 (PGE2). We demonstrated that 0.01 to 10 nM PGE2 protected dopaminergic neurons against LPS-induced neurotoxicity through a reduction of microglial release of pro-inflammatory factors in a dose-dependent manner. Mechanistically, neuroprotective effects elicited by PGE2 were mediated by the inhibition of microglial NOX2, a major superoxide-producing enzyme. This conclusion was supported by (1) the close relationship between inhibition of superoxide and PGE2-induced neuroprotective effects; (2) the mediation of PGE2-induced reduction of superoxide and neuroprotection via direct inhibition of the catalytic subunit of NOX2, gp91phox, rather than through the inhibition of conventional prostaglandin E2 receptors; and (3) abolishment of the neuroprotective effect of PGE2 in NOX2-deficient cultures. In summary, this study revealed a potential physiological role of PGE2 in maintaining brain immune homeostasis and protecting neurons via an EP receptor-independent mechanism.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.
| | - Yueh-Feng Sung
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Esteban A Oyarzabal
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Yu-Mei Tan
- U.S. Environmental Protection Agency, National Exposure Research Lab, Research Triangle Park, NC, USA
| | - Jeremy Leonard
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Mingri Guo
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Laboratory Medicine, Tianjin Haihe Hospital/Haihe Clinical Institute of Tianjin Medical University, Tianjin, China
| | - Shuo Li
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.,Department of Respiratory, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingshan Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Chun-Hsien Chu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA
| | - Shiou-Lan Chen
- Department of Neurology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ru-Band Lu
- Institute of Behavioral Medicine, College of Medicine & Hospital, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, National Cheng Kung University, Tainan, Taiwan
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, NIEHS/NIH, 111 T.W. Alexander Dr., Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
22
|
Anttila JE, Whitaker KW, Wires ES, Harvey BK, Airavaara M. Role of microglia in ischemic focal stroke and recovery: focus on Toll-like receptors. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:3-14. [PMID: 27389423 PMCID: PMC5214845 DOI: 10.1016/j.pnpbp.2016.07.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/20/2016] [Accepted: 07/02/2016] [Indexed: 12/21/2022]
Abstract
Stroke is the leading cause of disability in adults. Drug treatments that target stroke-induced pathological mechanisms and promote recovery are desperately needed. In the brain, an ischemic event triggers major inflammatory responses that are mediated by the resident microglial cells. In this review, we focus on the microglia activation after ischemic brain injury as a target of immunomodulatory therapeutics. We divide the microglia-mediated events following ischemic stroke into three categories: acute, subacute, and long-term events. This division encompasses the spatial and temporal dynamics of microglia as they participate in the pathophysiological changes that contribute to the symptoms and sequela of a stroke. The importance of Toll-like receptor (TLR) signaling in the outcomes of these pathophysiological changes is highlighted. Increasing evidence shows that microglia have a complex role in stroke pathophysiology, and they mediate both detrimental and beneficial effects on stroke outcome. So far, most of the pharmacological studies in experimental models of stroke have focused on neuroprotective strategies which are impractical for clinical applications. Post-ischemic inflammation is long lasting and thus, could provide a therapeutic target for novel delayed drug treatment. However, more studies are needed to elucidate the role of microglia in the recovery process from an ischemic stroke and to evaluate the therapeutic potential of modulating post-ischemic inflammation to promote functional recovery.
Collapse
Affiliation(s)
- Jenni E Anttila
- Institute of Biotechnology, P.O. Box 56, 00014, University of Helsinki, Finland
| | - Keith W Whitaker
- Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, USA; Human Research and Engineering Directorate, US Army Research Laboratory, Aberdeen, Proving Ground, MD 21005, USA
| | - Emily S Wires
- Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, USA
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, USA
| | - Mikko Airavaara
- Institute of Biotechnology, P.O. Box 56, 00014, University of Helsinki, Finland.
| |
Collapse
|
23
|
Abdyazdani N, Nourazarian A, Nozad Charoudeh H, Kazemi M, Feizy N, Akbarzade M, Mehdizadeh A, Rezaie J, Rahbarghazi R. The role of morphine on rat neural stem cells viability, neuro-angiogenesis and neuro-steroidgenesis properties. Neurosci Lett 2017; 636:205-212. [PMID: 27845244 DOI: 10.1016/j.neulet.2016.11.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 11/16/2022]
Abstract
A lack of comprehensive data exists on the effect of morphine on neural stem cell neuro-steroidogenesis and neuro-angiogenesis properties. We, herein, investigated the effects of morphine (100μM), naloxone (100μM) and their combination on rat neural stem cells viability, clonogenicity and Ki-67 expression over a period of 72h. Any alterations in the total fatty acids profile under treatment protocols were elucidated by direct transesterification method. We also monitored the expression of p53, aromatase and 5-alpha reductase by real-time PCR assay. To examine angiogenic capacity, in vitro tubulogenesis and the level of VE-cadherin transcript were investigated during neural to endothelial differentiation under the experimental procedure. Cells supplemented with morphine displayed reduced survival (p<0.01) and clonogenicity (p<0.001). Flow cytometric analysis showed a decrease in Ki-67 during 72h. Naloxone potentially blunted morphine-induced all effects. The normal levels of fatty acids, including saturated and unsaturated were altered by naloxone and morphine supplements. Following 48h, the up-regulation of p53, aromatase and 5-alpha reductase genes occurred in morphine-primed cells. Using three-dimensional culture models of angiogenesis and real time PCR assay, we showed morphine impaired the tubulogenesis properties of neural stem cells (p<0.001) by the inhibition of trans-differentiation into vascular cells and led to decrease of in VE-cadherin expression. Collectively, morphine strongly impaired the healthy status of neural stem cells by inducing p53 and concurrent elevation of aromatase and 5-alpha reductase activities especially during early 48h. Also, neural stem cells-being exposed to morphine lost their potency to elicit angiogenesis.
Collapse
Affiliation(s)
- Nima Abdyazdani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Masoumeh Kazemi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Feizy
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Akbarzade
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Cifuentes-Pagano ME, Meijles DN, Pagano PJ. Nox Inhibitors & Therapies: Rational Design of Peptidic and Small Molecule Inhibitors. Curr Pharm Des 2016; 21:6023-35. [PMID: 26510437 DOI: 10.2174/1381612821666151029112013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
Oxidative stress-related diseases underlie many if not all of the major leading causes of death in United States and the Western World. Thus, enormous interest from both academia and pharmaceutical industry has been placed on the development of agents which attenuate oxidative stress. With that in mind, great efforts have been placed in the development of inhibitors of NADPH oxidase (Nox), the major enzymatic source of reactive oxygen species and oxidative stress in many cells and tissue. The regulation of a catalytically active Nox enzyme involves numerous protein-protein interactions which, in turn, afford numerous targets for inhibition of its activity. In this review, we will provide an updated overview of the available Nox inhibitors, both peptidic and small molecules, and discuss the body of data related to their possible mechanisms of action and specificity towards each of the various isoforms of Nox. Indeed, there have been some very notable successes. However, despite great commitment by many in the field, the need for efficacious and well-characterized, isoform-specific Nox inhibitors, essential for the treatment of major diseases as well as for delineating the contribution of a given Nox in physiological redox signalling, continues to grow.
Collapse
Affiliation(s)
| | | | - Patrick J Pagano
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Biomedical Science Tower, 12th Floor, Room E1247, 200 Lothrop St., Pittsburgh, PA 15261, USA.
| |
Collapse
|
25
|
Blakely PK, Huber AK, Irani DN. Type-1 angiotensin receptor signaling in central nervous system myeloid cells is pathogenic during fatal alphavirus encephalitis in mice. J Neuroinflammation 2016; 13:196. [PMID: 27562117 PMCID: PMC5000512 DOI: 10.1186/s12974-016-0683-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/18/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.
Collapse
Affiliation(s)
- Pennelope K Blakely
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Room 4007, A. Alfred Taubman Biomedical Sciences Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Amanda K Huber
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Room 4007, A. Alfred Taubman Biomedical Sciences Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - David N Irani
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School, Room 4007, A. Alfred Taubman Biomedical Sciences Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
26
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
27
|
MA YI, GONG XUN, MO YINGLI, WU SAIZHU. Polydatin inhibits the oxidative stress-induced proliferation of vascular smooth muscle cells by activating the eNOS/SIRT1 pathway. Int J Mol Med 2016; 37:1652-60. [DOI: 10.3892/ijmm.2016.2554] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 03/28/2016] [Indexed: 11/06/2022] Open
|
28
|
Taylor AMW, Castonguay A, Ghogha A, Vayssiere P, Pradhan AAA, Xue L, Mehrabani S, Wu J, Levitt P, Olmstead MC, De Koninck Y, Evans CJ, Cahill CM. Neuroimmune Regulation of GABAergic Neurons Within the Ventral Tegmental Area During Withdrawal from Chronic Morphine. Neuropsychopharmacology 2016; 41:949-59. [PMID: 26202104 PMCID: PMC4748420 DOI: 10.1038/npp.2015.221] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 12/13/2022]
Abstract
Opioid dependence is accompanied by neuroplastic changes in reward circuitry leading to a negative affective state contributing to addictive behaviors and risk of relapse. The current study presents a neuroimmune mechanism through which chronic opioids disrupt the ventral tegmental area (VTA) dopaminergic circuitry that contributes to impaired reward behavior. Opioid dependence was induced in rodents by treatment with escalating doses of morphine. Microglial activation was observed in the VTA following spontaneous withdrawal from chronic morphine treatment. Opioid-induced microglial activation resulted in an increase in brain-derived neurotrophic factor (BDNF) expression and a reduction in the expression and function of the K(+)Cl(-) co-transporter KCC2 within VTA GABAergic neurons. Inhibition of microglial activation or interfering with BDNF signaling prevented the loss of Cl(-) extrusion capacity and restored the rewarding effects of cocaine in opioid-dependent animals. Consistent with a microglial-derived BDNF-induced disruption of reward, intra-VTA injection of BDNF or a KCC2 inhibitor resulted in a loss of cocaine-induced place preference in opioid-naïve animals. The loss of the extracellular Cl(-) gradient undermines GABAA-mediated inhibition, and represents a mechanism by which chronic opioid treatments can result in blunted reward circuitry. This study directly implicates microglial-derived BDNF as a negative regulator of reward in opioid-dependent states, identifying new therapeutic targets for opiate addictive behaviors.
Collapse
Affiliation(s)
- Anna M W Taylor
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, CA, USA
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Annie Castonguay
- Institut Universitaire en Santé Mentale de Québec, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Atefeh Ghogha
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Pia Vayssiere
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Amynah A A Pradhan
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Lihua Xue
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sadaf Mehrabani
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Juli Wu
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Children's Hospital Los Angeles and the Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Mary C Olmstead
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Christopher J Evans
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Catherine M Cahill
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, CA, USA
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
29
|
Haslund-Vinding J, McBean G, Jaquet V, Vilhardt F. NADPH oxidases in oxidant production by microglia: activating receptors, pharmacology and association with disease. Br J Pharmacol 2016; 174:1733-1749. [PMID: 26750203 DOI: 10.1111/bph.13425] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/15/2015] [Accepted: 01/07/2016] [Indexed: 12/26/2022] Open
Abstract
Microglia are the resident immune cells of the CNS and constitute a self-sustaining population of CNS-adapted tissue macrophages. As mononuclear phagocytic cells, they express high levels of superoxide-producing NADPH oxidases (NOX). The sole function of the members of the NOX family is to generate reactive oxygen species (ROS) that are believed to be important in CNS host defence and in the redox signalling circuits that shape the different activation phenotypes of microglia. NOX are also important in pathological conditions, where over-generation of ROS contributes to neuronal loss via direct oxidative tissue damage or disruption of redox signalling circuits. In this review, we assess the evidence for involvement of NOX in CNS physiopathology, with particular emphasis on the most important surface receptors that lead to generation of NOX-derived ROS. We evaluate the potential significance of the subcellular distribution of NOX isoforms for redox signalling or release of ROS to the extracellular medium. Inhibitory mechanisms that have been reported to restrain NOX activity in microglia and macrophages in vivo are also discussed. We provide a critical appraisal of frequently used and recently developed NOX inhibitors. Finally, we review the recent literature on NOX and other sources of ROS that are involved in activation of the inflammasome and discuss the potential influence of microglia-derived oxidants on neurogenesis, neural differentiation and culling of surplus progenitor cells. The degree to which excessive, badly timed or misplaced NOX activation in microglia may affect neuronal homeostasis in physiological or pathological conditions certainly merits further investigation. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- J Haslund-Vinding
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark.,Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - G McBean
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Ireland
| | - V Jaquet
- Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - F Vilhardt
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
30
|
Wang X, Zhang Y, Peng Y, Hutchinson MR, Rice KC, Yin H, Watkins LR. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol 2016; 173:856-69. [PMID: 26603732 DOI: 10.1111/bph.13394] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The toll-like receptor TLR4 is involved in neuropathic pain and in drug reward and reinforcement. The opioid inactive isomers (+)-naltrexone and (+)-naloxone act as TLR4 antagonists, reversing neuropathic pain and reducing opioid and cocaine reward and reinforcement. However, how these agents modulate TLR4 signalling is not clear. Here, we have elucidated the molecular mechanism of (+)-naltrexone and (+)-naloxone on TLR4 signalling. EXPERIMENTAL APPROACH BV-2 mouse microglial cell line, primary rat microglia and primary rat peritoneal macrophages were treated with LPS and TLR4 signalling inhibitors. Effects were measured using Western blotting, luciferase reporter assays, fluorescence microscopy and ELISA KEY RESULTS: (+)-Naltrexone and (+)-naloxone were equi-potent inhibitors of the LPS-induced TLR4 downstream signalling and induction of the pro-inflammatory factors NO and TNF-α. Similarly, (+)-naltrexone or (+)-naloxone inhibited production of reactive oxygen species and increased microglial phagocytosis, induced by LPS. However, (+)-naltrexone and (+)-naloxone did not directly inhibit the increased production of IL-1β, induced by LPS. The drug interaction of (+)-naloxone and (+)-naltrexone was additive. (+)-Naltrexone or (+)-naloxone inhibited LPS-induced activation of IFN regulatory factor 3 and production of IFN-β. However, they did not inhibit TLR4 signalling via the activation of either NF-κB, p38 or JNK in these cellular models. CONCLUSIONS AND IMPLICATIONS (+)-Naltrexone and (+)-naloxone were TRIF-IFN regulatory factor 3 axis-biased TLR4 antagonists. They blocked TLR4 downstream signalling leading to NO, TNF-α and reactive oxygen species. This pattern may explain, at least in part, the in vivo therapeutic effects of (+)-naltrexone and (+)-naloxone.
Collapse
Affiliation(s)
- X Wang
- Chemical Biology Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Y Zhang
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Y Peng
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - M R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - K C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20892, USA
| | - H Yin
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - L R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| |
Collapse
|
31
|
Morphine Inhibited the Rat Neural Stem Cell Proliferation Rate by Increasing Neuro Steroid Genesis. Neurochem Res 2016; 41:1410-9. [DOI: 10.1007/s11064-016-1847-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/28/2015] [Accepted: 01/22/2016] [Indexed: 11/26/2022]
|
32
|
Hota D, Srinivasan A, Dutta P, Bhansali A, Chakrabarti A. Off-Label, Low-Dose Naltrexone for Refractory Painful Diabetic Neuropathy. PAIN MEDICINE 2015; 17:790-1. [PMID: 26814245 DOI: 10.1093/pm/pnv009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/05/2015] [Indexed: 11/14/2022]
Affiliation(s)
- Debasish Hota
- *Department of Pharmacology, All India Institute of Medical Sciences, Bhubaneswar, India
| | | | - Pinaki Dutta
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
33
|
Hirano K, Chen WS, Chueng ALW, Dunne AA, Seredenina T, Filippova A, Ramachandran S, Bridges A, Chaudry L, Pettman G, Allan C, Duncan S, Lee KC, Lim J, Ma MT, Ong AB, Ye NY, Nasir S, Mulyanidewi S, Aw CC, Oon PP, Liao S, Li D, Johns DG, Miller ND, Davies CH, Browne ER, Matsuoka Y, Chen DW, Jaquet V, Rutter AR. Discovery of GSK2795039, a Novel Small Molecule NADPH Oxidase 2 Inhibitor. Antioxid Redox Signal 2015; 23:358-74. [PMID: 26135714 PMCID: PMC4545375 DOI: 10.1089/ars.2014.6202] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS The NADPH oxidase (NOX) family of enzymes catalyzes the formation of reactive oxygen species (ROS). NOX enzymes not only have a key role in a variety of physiological processes but also contribute to oxidative stress in certain disease states. To date, while numerous small molecule inhibitors have been reported (in particular for NOX2), none have demonstrated inhibitory activity in vivo. As such, there is a need for the identification of improved NOX inhibitors to enable further evaluation of the biological functions of NOX enzymes in vivo as well as the therapeutic potential of NOX inhibition. In this study, both the in vitro and in vivo pharmacological profiles of GSK2795039, a novel NOX2 inhibitor, were characterized in comparison with other published NOX inhibitors. RESULTS GSK2795039 inhibited both the formation of ROS and the utilization of the enzyme substrates, NADPH and oxygen, in a variety of semirecombinant cell-free and cell-based NOX2 assays. It inhibited NOX2 in an NADPH competitive manner and was selective over other NOX isoforms, xanthine oxidase, and endothelial nitric oxide synthase enzymes. Following systemic administration in mice, GSK2795039 abolished the production of ROS by activated NOX2 enzyme in a paw inflammation model. Furthermore, GSK2795039 showed activity in a murine model of acute pancreatitis, reducing the levels of serum amylase triggered by systemic injection of cerulein. INNOVATION AND CONCLUSIONS GSK2795039 is a novel NOX2 inhibitor that is the first small molecule to demonstrate inhibition of the NOX2 enzyme in vivo.
Collapse
Affiliation(s)
- Kazufumi Hirano
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Woei Shin Chen
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Adeline L W Chueng
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Angela A Dunne
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Tamara Seredenina
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - Aleksandra Filippova
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - Sumitra Ramachandran
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Angela Bridges
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Laiq Chaudry
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Gary Pettman
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Craig Allan
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Sarah Duncan
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Kiew Ching Lee
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Jean Lim
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - May Thu Ma
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Agnes B Ong
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Nicole Y Ye
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Shabina Nasir
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Sri Mulyanidewi
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Chiu Cheong Aw
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Pamela P Oon
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Shihua Liao
- 4 Neuroimmunology Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Shanghai, China
| | - Dizheng Li
- 4 Neuroimmunology Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Shanghai, China
| | - Douglas G Johns
- 5 Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline , King of Prussia, Pennsylvania
| | - Neil D Miller
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Ceri H Davies
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Edward R Browne
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Yasuji Matsuoka
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Deborah W Chen
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Vincent Jaquet
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - A Richard Rutter
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| |
Collapse
|
34
|
Substance P enhances microglial density in the substantia nigra through neurokinin-1 receptor/NADPH oxidase-mediated chemotaxis in mice. Clin Sci (Lond) 2015. [PMID: 26223840 DOI: 10.1042/cs20150008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The distribution of microglia varies greatly throughout the brain. The substantia nigra (SN) contains the highest density of microglia among different brain regions. However, the mechanism underlying this uneven distribution remains unclear. Substance P (SP) is a potent proinflammatory neuropeptide with high concentrations in the SN. We recently demonstrated that SP can regulate nigral microglial activity. In the present study, we further investigated the involvement of SP in modulating nigral microglial density in postnatal developing mice. Nigral microglial density was quantified in wild-type (WT) and SP-deficient mice from postnatal day 1 (P1) to P30. SP was detected at high levels in the SN as early as P1 and microglial density did not peak until around P30 in WT mice. SP-deficient mice (TAC1(-/-)) had a significant reduction in nigral microglial density. No differences in the ability of microglia to proliferate were observed between TAC1(-/-) and WT mice, suggesting that SP may alter microglial density through chemotaxic recruitment. SP was confirmed to dose-dependently attract microglia using a trans-well culture system. Mechanistic studies revealed that both the SP receptor neurokinin-1 receptor (NK1R) and the superoxide-producing enzyme NADPH oxidase (NOX2) were necessary for SP-mediated chemotaxis in microglia. Furthermore, genetic ablation and pharmacological inhibition of NK1R or NOX2 attenuated SP-induced microglial migration. Finally, protein kinase Cδ (PKCδ) was recognized to couple SP/NK1R-mediated NOX2 activation. Altogether, we found that SP partly accounts for the increased density of microglia in the SN through chemotaxic recruitment via a novel NK1R-NOX2 axis-mediated pathway.
Collapse
|
35
|
Jiang L, Chen SH, Chu CH, Wang SJ, Oyarzabal E, Wilson B, Sanders V, Xie K, Wang Q, Hong JS. A novel role of microglial NADPH oxidase in mediating extra-synaptic function of norepinephrine in regulating brain immune homeostasis. Glia 2015; 63:1057-72. [PMID: 25740080 DOI: 10.1002/glia.22801] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 01/15/2015] [Indexed: 12/30/2022]
Abstract
Although the peripheral anti-inflammatory effect of norepinephrine (NE) is well documented, the mechanism by which this neurotransmitter functions as an anti-inflammatory/neuroprotective agent in the central nervous system (CNS) is unclear. This article aimed to determine the anti-inflammatory/neuroprotective effects and underlying mechanisms of NE in inflammation-based dopaminergic neurotoxicity models. In mice, NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected at 6 months of lipopolysaccharide (LPS)-induced neuroinflammation. It was found that NE depletion enhanced LPS-induced dopaminergic neuron loss in the substantia nigra. This piece of in vivo data prompted us to conduct a series of studies in an effort to elucidate the mechanism as to how NE affects dopamine neuron survival by using primary midbrain neuron/glia cultures. Results showed that submicromolar concentrations of NE dose-dependently protected dopaminergic neurons from LPS-induced neurotoxicity by inhibiting microglia activation and subsequent release of pro-inflammatory factors. However, NE-elicited neuroprotection was not totally abolished in cultures from β2-adrenergic receptor (β2-AR)-deficient mice, suggesting that novel pathways other than β2-AR are involved. To this end, It was found that submicromolar NE dose-dependently inhibited NADPH oxidase (NOX2)-generated superoxide, which contributes to the anti-inflammatory and neuroprotective effects of NE. This novel mechanism was indeed adrenergic receptors independent since both (+) and (-) optic isomers of NE displayed the same potency. We further demonstrated that NE inhibited LPS-induced NOX2 activation by blocking the translocation of its cytosolic subunit to plasma membranes. In summary, we revealed a potential physiological role of NE in maintaining brain immune homeostasis and protecting neurons via a novel mechanism.
Collapse
Affiliation(s)
- Lulu Jiang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China; Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, 27709
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Substance P exacerbates dopaminergic neurodegeneration through neurokinin-1 receptor-independent activation of microglial NADPH oxidase. J Neurosci 2015; 34:12490-503. [PMID: 25209287 DOI: 10.1523/jneurosci.2238-14.2014] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although dysregulated substance P (SP) has been implicated in the pathophysiology of Parkinson's disease (PD), how SP affects the survival of dopaminergic neurons remains unclear. Here, we found that mice lacking endogenous SP (TAC1(-/-)), but not those deficient in the SP receptor (neurokinin-1 receptor, NK1R), were more resistant to lipopolysaccharide (LPS)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigral dopaminergic neurodegeneration than wild-type controls, suggesting a NK1R-independent toxic action of SP. In vitro dose-response studies revealed that exogenous SP enhanced LPS- and 1-methyl-4-phenylpyridinium (MPP(+))-induced dopaminergic neurodegeneration in a bimodal manner, peaking at submicromolar and subpicomolar concentrations, but was substantially less effective at intermediate concentrations. Mechanistically, the actions of submicromolar levels of SP were NK1R-dependent, whereas subpicomolar SP-elicited actions required microglial NADPH oxidase (NOX2), the key superoxide-producing enzyme, but not NK1R. Subpicomolar concentrations of SP activated NOX2 by binding to the catalytic subunit gp91(phox) and inducing membrane translocation of the cytosolic subunits p47(phox) and p67(phox). The importance of NOX2 was further corroborated by showing that inhibition or disruption of NOX2 blocked subpicomolar SP-exacerbated neurotoxicity. Together, our findings revealed a critical role of microglial NOX2 in mediating the neuroinflammatory and dopaminergic neurodegenerative effects of SP, which may provide new insights into the pathogenesis of PD.
Collapse
|
37
|
Brandes RP, Weissmann N, Schröder K. Nox family NADPH oxidases: Molecular mechanisms of activation. Free Radic Biol Med 2014; 76:208-26. [PMID: 25157786 DOI: 10.1016/j.freeradbiomed.2014.07.046] [Citation(s) in RCA: 518] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 11/21/2022]
Abstract
NADPH oxidases of the Nox family are important enzymatic sources of reactive oxygen species (ROS). Numerous homologue-specific mechanisms control the activity of this enzyme family involving calcium, free fatty acids, protein-protein interactions, intracellular trafficking, and posttranslational modifications such as phosphorylation, acetylation, or sumoylation. After a brief review on the classic pathways of Nox activation, this article will focus on novel mechanisms of homologue-specific activity control and on cell-specific aspects which govern Nox activity. From these findings of the recent years it must be concluded that the activity control of Nox enzymes is much more complex than anticipated. Moreover, depending on the cellular activity state, Nox enzymes are selectively activated or inactivated. The complex upstream signaling aspects of these events make the development of "intelligent" Nox inhibitors plausible, which selectively attenuate disease-related Nox-mediated ROS formation without altering physiological signaling ROS. This approach might be of relevance for Nox-mediated tissue injury in ischemia-reperfusion and inflammation and also for chronic Nox overactivation as present in cancer initiation and cardiovascular disease.
Collapse
Affiliation(s)
- Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany.
| | - Norbert Weissmann
- ECCPS, Justus-Liebig-Universität, Member of the DZL, Giessen, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany
| |
Collapse
|
38
|
Dibutyltin promotes oxidative stress and increases inflammatory mediators in BV-2 microglia cells. Toxicol Lett 2014; 230:177-87. [DOI: 10.1016/j.toxlet.2014.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/29/2014] [Accepted: 03/04/2014] [Indexed: 01/24/2023]
|
39
|
In vivo veritas: (+)-Naltrexone's actions define translational importance. Trends Pharmacol Sci 2014; 35:432-3. [DOI: 10.1016/j.tips.2014.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/08/2014] [Indexed: 12/26/2022]
|
40
|
Wang Q, Chu CH, Oyarzabal E, Jiang L, Chen SH, Wilson B, Qian L, Hong JS. Subpicomolar diphenyleneiodonium inhibits microglial NADPH oxidase with high specificity and shows great potential as a therapeutic agent for neurodegenerative diseases. Glia 2014; 62:2034-43. [PMID: 25043383 DOI: 10.1002/glia.22724] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/11/2014] [Accepted: 07/03/2014] [Indexed: 01/22/2023]
Abstract
Activation of microglial NADPH oxidase (NOX2) plays a critical role in mediating neuroinflammation, which is closely linked with the pathogenesis of a variety of neurodegenerative diseases, including Parkinson's disease (PD). The inhibition of NOX2-generated superoxide has become an effective strategy for developing disease-modifying therapies for PD. However, the lack of specific and potent NOX2 inhibitors has hampered the progress of this approach. Diphenyleneiodonium (DPI) is a widely used, long-acting NOX2 inhibitor. However, due to its non-specificity for NOX2 and high cytotoxicity at standard doses (µM), DPI has been precluded from human studies. In this study, using ultra-low doses of DPI, we aimed to: (1) investigate whether these problems could be circumvented and (2) determine whether ultra-low doses of DPI were able to preserve its utility as a potent NOX2 inhibitor. We found that DPI at subpicomolar concentrations (10(-14) and 10(-13) M) displays no toxicity in primary midbrain neuron-glia cultures. More importantly, we observed that subpicomolar DPI inhibited phorbol myristate acetate (PMA)-induced activation of NOX2. The same concentrations of DPI did not inhibit the activities of a series of flavoprotein-containing enzymes. Furthermore, potent neuroprotective efficacy was demonstrated in a post-treatment study. When subpicomolar DPI was added to neuron-glia cultures pretreated with lipopolysaccharide, 1-methyl-4-phenylpyridinium or rotenone, it potently protected the dopaminergic neurons. In summary, DPI's unique combination of high specificity toward NOX2, low cytotoxicity and potent neuroprotective efficacy in post-treatment regimens suggests that subpicomolar DPI may be an ideal candidate for further animal studies and potential clinical trials.
Collapse
Affiliation(s)
- Qingshan Wang
- Neuropharmacology Section, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cifuentes-Pagano E, Meijles DN, Pagano PJ. The quest for selective nox inhibitors and therapeutics: challenges, triumphs and pitfalls. Antioxid Redox Signal 2014; 20:2741-54. [PMID: 24070014 PMCID: PMC4026400 DOI: 10.1089/ars.2013.5620] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Numerous studies in animal models and human subjects corroborate that elevated levels of reactive oxygen species (ROS) play a pivotal role in the progression of multiple diseases. As a major source of ROS in many organ systems, the NADPH oxidase (Nox) has become a prime target for therapeutic development. RECENT ADVANCES In recent years, intense efforts have been dedicated to the development of pan- and isoform-specific Nox inhibitors as opposed to antioxidants that proved ineffective in clinical trials. Over the past decade, an array of compounds has been proposed in an attempt to fill this void. CRITICAL ISSUES Although many of these compounds have proven effective as Nox enzyme family inhibitors, isoform specificity has posed a formidable challenge to the scientific community. This review surveys the most prominent Nox inhibitors, and discusses potential isoform specificity, known mechanisms of action, and shortcomings. Some of these inhibitors hold substantial promise as targeted therapeutics. FUTURE DIRECTIONS Increased insight into the mechanisms of action and regulation of this family of enzymes as well as atomic structures of key Nox subunits are expected to give way to a broader spectrum of more potent, efficacious, and specific molecules. These lead molecules will assuredly serve as a basis for drug development aimed at treating a wide array of diseases associated with increased Nox activity.
Collapse
Affiliation(s)
- Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | | | | |
Collapse
|
42
|
Younger J, Parkitny L, McLain D. The use of low-dose naltrexone (LDN) as a novel anti-inflammatory treatment for chronic pain. Clin Rheumatol 2014; 33:451-9. [PMID: 24526250 PMCID: PMC3962576 DOI: 10.1007/s10067-014-2517-2] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 01/22/2014] [Accepted: 01/26/2014] [Indexed: 02/08/2023]
Abstract
Low-dose naltrexone (LDN) has been demonstrated to reduce symptom severity in conditions such as fibromyalgia, Crohn's disease, multiple sclerosis, and complex regional pain syndrome. We review the evidence that LDN may operate as a novel anti-inflammatory agent in the central nervous system, via action on microglial cells. These effects may be unique to low dosages of naltrexone and appear to be entirely independent from naltrexone's better-known activity on opioid receptors. As a daily oral therapy, LDN is inexpensive and well-tolerated. Despite initial promise of efficacy, the use of LDN for chronic disorders is still highly experimental. Published trials have low sample sizes, and few replications have been performed. We cover the typical usage of LDN in clinical trials, caveats to using the medication, and recommendations for future research and clinical work. LDN may represent one of the first glial cell modulators to be used for the management of chronic pain disorders.
Collapse
|
43
|
Yang CP, Cherng CH, Wu CT, Huang HY, Tao PL, Lee SO, Wong CS. Intrathecal ultra-low dose naloxone enhances the antihyperalgesic effects of morphine and attenuates tumor necrosis factor-α and tumor necrosis factor-α receptor 1 expression in the dorsal horn of rats with partial sciatic nerve transection. Anesth Analg 2014; 117:1493-502. [PMID: 24257399 DOI: 10.1213/ane.0000000000000020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Glutamate homeostasis and microglia activation play an important role in the development and maintenance of neuropathic pain. We designed this investigation to examine whether ultra-low dose naloxone administered alone or in combination with morphine could alter the concentration of the excitatory amino acids (EAAs) glutamate and aspartate, as well as the expression of tumor necrosis factor-α (TNF-α) and its receptors (TNFR1 and TNFR2) in the spinal cord dorsal horn of rats with partial sciatic nerve transection (PST). METHODS Male Wistar rats underwent intrathecal catheter implantation for drug delivery and were divided in 7 groups: sham-operated + saline (sham), PST + saline (S), PST + 15 ng naloxone (n), PST + 15 µg naloxone (N), PST + 10 µg morphine (M), PST + 15 ng naloxone + 10 µg morphine (Mn), PST + 15 µg naloxone + 10 µg morphine (MN). Thermal withdrawal latency and mechanical withdrawal threshold, TNF-α and TNFR expression in the spinal cord and dorsal root ganglia, and EAAs glutamate and aspartate concentration in cerebrospinal fluid dialysates were measured. RESULTS Ten days after PST, rats developed hyperalgesia (P < 0.0001) and allodynia (P < 0.0001), and increased TNF-α (P < 0.0001) and TNFR1 expression (P = 0.0009) were measured in the ipsilateral spinal cord dorsal horn. The antihyperalgesic and antiallodynic effects of morphine (10 μg) were abolished by high-dose naloxone (15 μg; P = 0.0031) but enhanced by ultra-low dose naloxone (15 ng; P = 0.0015), and this was associated with a reduction of TNF-α (P < 0.0001) and TNFR1 (P = 0.0009) expression in the spinal cord dorsal horn and EAAs concentration (glutamate: P = 0.0001; aspartate: P = 0.004) in cerebrospinal fluid dialysate. Analysis of variance (ANOVA) or Student t test with Bonferroni correction were used for statistical analysis. CONCLUSIONS Ultra-low dose naloxone enhances the antihyperalgesia and antiallodynia effects of morphine in PST rats, possibly by reducing TNF-α and TNFR1 expression, and EAAs concentrations in the spinal dorsal horn. Ultra-low dose naloxone may be a useful adjuvant for increasing the analgesic effect of morphine in neuropathic pain conditions.
Collapse
Affiliation(s)
- Chih-Ping Yang
- From the *Division of Anesthesiology, Armed Forces Taoyuan General Hospital, Taoyuan; †Tri-Service General Hospital, ‡Department of Anesthesiology, and §Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei; ‖Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County; and ¶Department of Anesthesiology, Cathy General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
45
|
Won SY, Kim SR, Maeng S, Jin BK. Interleukin-13/Interleukin-4-induced oxidative stress contributes to death of prothrombinkringle-2 (pKr-2)-activated microglia. J Neuroimmunol 2013; 265:36-42. [PMID: 24090651 DOI: 10.1016/j.jneuroim.2013.09.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/12/2013] [Accepted: 09/14/2013] [Indexed: 01/28/2023]
Abstract
The present study examined whether Interleukin-13 (IL-13) or IL-4, an anti-inflammatory cytokine, could induce cell death of activated microglia by prothrombin kringle-2 (pKr-2) which is a domain of prothrombin distinct from thrombin. Microglia cell death was detected at eight days after co-treatment of pKr-2 with IL-13/IL-4 in vitro. This cell death was assessed by live assay, dead assay, TUNEL and MTT assay. In parallel, reactive oxygen species (ROS) production was evident as assessed by superoxide assay, WST-1 and analyzing DCF in combination of pKr-2 and IL-13 or IL-4 treated microglia. The IL-13/IL-4-enhanced ROS production and cell death in pKr-2 activated microglia was partially inhibited by an NADPH oxidase inhibitor, apocynin and/or by several antioxidants. Moreover, Western blot analysis showed a significant increase in cyclooxygenase-2 (COX-2) expression in combination of pKr-2 and IL-13 or IL-4 treated microglia, which was partially inhibited by apocynin and an antioxidant, trolox. Additional studies demonstrated that microglia cell death was reversed by treatment with COX-2 inhibitor, NS398. Our data strongly suggest that oxidative stress and COX-2 activation through NADPH oxidase may contribute to IL-13/IL-4 induced cell death of pKr-2 activated microglia.
Collapse
Affiliation(s)
- So-Yoon Won
- Department of Biochemistry and Medical Research Center, Chungbuk National University College of Medicine, Cheongju 361-763, South Korea
| | | | | | | |
Collapse
|
46
|
Zhao X, Bian Y, Sun Y, Li L, Wang L, Zhao C, Shen Y, Song Q, Qu Y, Niu S, Wu W, Gao F. Effects of moderate exercise over different phases on age-related physiological dysfunction in testes of SAMP8 mice. Exp Gerontol 2013; 48:869-80. [DOI: 10.1016/j.exger.2013.05.063] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 04/26/2013] [Accepted: 05/30/2013] [Indexed: 02/02/2023]
|
47
|
Filer CN. Morphinan alkaloids labeled with tritium: synthesis and applications. J Labelled Comp Radiopharm 2013; 56:639-48. [DOI: 10.1002/jlcr.3094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Crist N. Filer
- PerkinElmer Life Sciences & Technology, Inc.; 940 Winter Street Waltham MA 02451 USA
| |
Collapse
|
48
|
Rodiño-Janeiro BK, Paradela-Dobarro B, Castiñeiras-Landeira MI, Raposeiras-Roubín S, González-Juanatey JR, Álvarez E. Current status of NADPH oxidase research in cardiovascular pharmacology. Vasc Health Risk Manag 2013; 9:401-28. [PMID: 23983473 PMCID: PMC3750863 DOI: 10.2147/vhrm.s33053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The implications of reactive oxygen species in cardiovascular disease have been known for some decades. Rationally, therapeutic antioxidant strategies combating oxidative stress have been developed, but the results of clinical trials have not been as good as expected. Therefore, to move forward in the design of new therapeutic strategies for cardiovascular disease based on prevention of production of reactive oxygen species, steps must be taken on two fronts, ie, comprehension of reduction-oxidation signaling pathways and the pathophysiologic roles of reactive oxygen species, and development of new, less toxic, and more selective nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors, to clarify both the role of each NADPH oxidase isoform and their utility in clinical practice. In this review, we analyze the value of NADPH oxidase as a therapeutic target for cardiovascular disease and the old and new pharmacologic agents or strategies to prevent NADPH oxidase activity. Some inhibitors and different direct or indirect approaches are available. Regarding direct NADPH oxidase inhibition, the specificity of NADPH oxidase is the focus of current investigations, whereas the chemical structure-activity relationship studies of known inhibitors have provided pharmacophore models with which to search for new molecules. From a general point of view, small-molecule inhibitors are preferred because of their hydrosolubility and oral bioavailability. However, other possibilities are not closed, with peptide inhibitors or monoclonal antibodies against NADPH oxidase isoforms continuing to be under investigation as well as the ongoing search for naturally occurring compounds. Likewise, some different approaches include inhibition of assembly of the NADPH oxidase complex, subcellular translocation, post-transductional modifications, calcium entry/release, electron transfer, and genetic expression. High-throughput screens for any of these activities could provide new inhibitors. All this knowledge and the research presently underway will likely result in development of new drugs for inhibition of NADPH oxidase and application of therapeutic approaches based on their action, for the treatment of cardiovascular disease in the next few years.
Collapse
Affiliation(s)
- Bruno K Rodiño-Janeiro
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- European Molecular Biology Laboratory, Grenoble, France
| | | | | | - Sergio Raposeiras-Roubín
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
| | - José R González-Juanatey
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| | - Ezequiel Álvarez
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| |
Collapse
|
49
|
Hutchinson MR, Watkins LR. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 2013; 76 Pt B:218-27. [PMID: 23764149 DOI: 10.1016/j.neuropharm.2013.05.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/13/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
A major development in drug addiction research in recent years has been the discovery that immune signaling within the central nervous system contributes significantly to mesolimbic dopamine reward signaling induced by drugs of abuse, and hence is involved in the presentation of reward behaviors. Additionally, in the case of opioids, these hypotheses have advanced through to the discovery of the novel site of opioid action at the innate immune pattern recognition receptor Toll-like receptor 4 as the necessary triggering event that engages this reward facilitating central immune signaling. Thus, the hypothesis of major proinflammatory contributions to drug abuse was born. This review will examine these key discoveries, but also address several key lingering questions of how central immune signaling is able to contribute in this fashion to the pharmacodynamics of drugs of abuse. It is hoped that by combining the collective wisdom of neuroscience, immunology and pharmacology, into Neuroimmunopharmacology, we may more fully understanding the neuronal and immune complexities of how drugs of abuse, such as opioids, create their rewarding and addiction states. Such discoveries will point us in the direction such that one day soon we might successfully intervene to successfully treat drug addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Level 5, Medical School South, Frome Rd, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
50
|
Theberge FR, Li X, Kambhampati S, Pickens CL, St Laurent R, Bossert JM, Baumann MH, Hutchinson MR, Rice KC, Watkins LR, Shaham Y. Effect of chronic delivery of the Toll-like receptor 4 antagonist (+)-naltrexone on incubation of heroin craving. Biol Psychiatry 2013; 73:729-37. [PMID: 23384483 PMCID: PMC3615146 DOI: 10.1016/j.biopsych.2012.12.019] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/10/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND Recent evidence implicates toll-like receptor 4 (TLR4) in opioid analgesia, tolerance, conditioned place preference, and self-administration. Here, we determined the effect of the TLR4 antagonist (+)-naltrexone (a μ-opioid receptor inactive isomer) on the time-dependent increases in cue-induced heroin seeking after withdrawal (incubation of heroin craving). METHODS In an initial experiment, we trained rats for 9 hours per day to self-administer heroin (.1 mg/kg/infusion) for 9 days; lever presses were paired with a 5-second tone-light cue. We then assessed cue-induced heroin seeking in 30-minute extinction sessions on withdrawal day 1; immediately after testing, we surgically implanted rats with Alzet minipumps delivering (+)-naltrexone (0, 7.5, 15, 30 mg/kg/day, subcutaneous) for 14 days. We then tested the rats for incubated cue-induced heroin seeking in 3-hour extinction tests on withdrawal day 13. RESULTS We found that chronic delivery of (+)-naltrexone via minipumps during the withdrawal phase decreased incubated cue-induced heroin seeking. In follow-up experiments, we found that acute injections of (+)-naltrexone immediately before withdrawal day 13 extinction tests had no effect on incubated cue-induced heroin seeking. Furthermore, chronic delivery of (+)-naltrexone (15 or 30 mg/kg/day) or acute systemic injections (15 or 30 mg/kg) had no effect on ongoing extended access heroin self-administration. Finally, in rats trained to self-administer methamphetamine (.1 mg/kg/infusion, 9 hours/day, 9 days), chronic delivery of (+)-naltrexone (30 mg/kg/day) during the withdrawal phase had no effect on incubated cue-induced methamphetamine seeking. CONCLUSIONS The present results suggest a critical role of TLR4 in the development of incubation of heroin, but not methamphetamine, craving.
Collapse
Affiliation(s)
- Florence R Theberge
- Intramural Research Program of the National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|