1
|
Huré JB, Foucault L, Ghayad LM, Marie C, Vachoud N, Baudouin L, Azmani R, Ivjanin N, Arevalo-Nuevo A, Pigache M, Bouslama-Oueghlani L, Chemelle JA, Dronne MA, Terreux R, Hassan B, Gueyffier F, Raineteau O, Parras C. Pharmacogenomic screening identifies and repurposes leucovorin and dyclonine as pro-oligodendrogenic compounds in brain repair. Nat Commun 2024; 15:9837. [PMID: 39537633 PMCID: PMC11561360 DOI: 10.1038/s41467-024-54003-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Oligodendrocytes are critical for CNS myelin formation and are involved in preterm-birth brain injury (PBI) and multiple sclerosis (MS), both of which lack effective treatments. We present a pharmacogenomic approach that identifies compounds with potent pro-oligodendrogenic activity, selected through a scoring strategy (OligoScore) based on their modulation of oligodendrogenic and (re)myelination-related transcriptional programs. Through in vitro neural and oligodendrocyte progenitor cell (OPC) cultures, ex vivo cerebellar explants, and in vivo mouse models of PBI and MS, we identify FDA-approved leucovorin and dyclonine as promising candidates. In a neonatal chronic hypoxia mouse model mimicking PBI, both compounds promote neural progenitor cell proliferation and oligodendroglial fate acquisition, with leucovorin further enhancing differentiation. In an adult MS model of focal de/remyelination, they improve lesion repair by promoting OPC differentiation while preserving the OPC pool. Additionally, they shift microglia from a pro-inflammatory to a pro-regenerative profile and enhance myelin debris clearance. These findings support the repurposing of leucovorin and dyclonine for clinical trials targeting myelin disorders, offering potential therapeutic avenues for PBI and MS.
Collapse
Affiliation(s)
- Jean-Baptiste Huré
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Louis Foucault
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Litsa Maria Ghayad
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Corentine Marie
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Nicolas Vachoud
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Lucas Baudouin
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Rihab Azmani
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Natalija Ivjanin
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alvaro Arevalo-Nuevo
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Morgane Pigache
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Lamia Bouslama-Oueghlani
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - Julie-Anne Chemelle
- Équipe ECMO, Laboratoire de Biologie Tissulaire et d'Ingénierie (LBTI), UMR5305, Lyon, France
| | - Marie-Aimée Dronne
- Claude Bernard University, UMR5558 Laboratoire de Biométrie et Biologie Evolutive, CNRS, Villeurbanne, France
| | - Raphaël Terreux
- Équipe ECMO, Laboratoire de Biologie Tissulaire et d'Ingénierie (LBTI), UMR5305, Lyon, France
| | - Bassem Hassan
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France
| | - François Gueyffier
- Claude Bernard University, UMR5558 Laboratoire de Biométrie et Biologie Evolutive, CNRS, Villeurbanne, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France.
| | - Carlos Parras
- Paris Brain Institute, Sorbonne Université, Inserm U1127, CNRS UMR 7225, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
2
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
3
|
Liguori F, Alberti F, Amadio S, Angelini DF, Pilesi E, Vitale G, Tesoriere G, Borsellino G, Vernì F, Volonté C. Pan-neuronal expression of human mutant SOD1 in Drosophila impairs survival and motor performance, induces early neuroinflammation and chromosome aberrations. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167192. [PMID: 38657911 DOI: 10.1016/j.bbadis.2024.167192] [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: 11/01/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Several mutations in the SOD1 gene encoding for the antioxidant enzyme Superoxide Dismutase 1, are associated with amyotrophic lateral sclerosis, a rare and devastating disease characterized by motor neuron degeneration and patients' death within 2-5 years from diagnosis. Motor neuron loss and related symptomatology manifest mostly in adult life and, to date, there is still a gap of knowledge on the precise cellular and molecular events preceding neurodegeneration. To deepen our awareness of the early phases of the disease, we leveraged two Drosophila melanogaster models pan-neuronally expressing either the mutation A4V or G85R of the human gene SOD1 (hSOD1A4V or hSOD1G85R). We demonstrate that pan-neuronal expression of the hSOD1A4V or hSOD1G85R pathogenic construct impairs survival and motor performance in transgenic flies. Moreover, protein and transcript analysis on fly heads indicates that mutant hSOD1 induction stimulates the glial marker Repo, up-regulates the IMD/Toll immune pathways through antimicrobial peptides and interferes with oxidative metabolism. Finally, cytological analysis of larval brains demonstrates hSOD1-induced chromosome aberrations. Of note, these parameters are found modulated in a timeframe when neurodegeneration is not detected. The novelty of our work is twofold: we have expressed for the first time hSOD1 mutations in all neurons of Drosophila and confirmed some ALS-related pathological phenotypes in these flies, confirming the power of SOD1 mutations in generating ALS-like phenotypes. Moreover, we have related SOD1 pathogenesis to chromosome aberrations and antimicrobial peptides up-regulation. These findings were unexplored in the SOD1-ALS field.
Collapse
Affiliation(s)
- Francesco Liguori
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy; Institute for Systems Analysis and Computer Science "Antonio Ruberti" (IASI), National Research Council (CNR), Via dei Taurini 19, 00185 Rome, Italy.
| | - Francesca Alberti
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Susanna Amadio
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Daniela Francesca Angelini
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Eleonora Pilesi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Giuseppe Vitale
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Giulia Tesoriere
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Giovanna Borsellino
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cinzia Volonté
- Experimental Neuroscience and Neurological Disease Models, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 65, 00143 Rome, Italy; Institute for Systems Analysis and Computer Science "Antonio Ruberti" (IASI), National Research Council (CNR), Via dei Taurini 19, 00185 Rome, Italy.
| |
Collapse
|
4
|
Chen H, Guo Z, Sun Y, Dai X. The immunometabolic reprogramming of microglia in Alzheimer's disease. Neurochem Int 2023; 171:105614. [PMID: 37748710 DOI: 10.1016/j.neuint.2023.105614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder (NDD). In the central nervous system (CNS), immune cells like microglia could reprogram intracellular metabolism to alter or exert cellular immune functions in response to environmental stimuli. In AD, microglia could be activated and differentiated into pro-inflammatory or anti-inflammatory phenotypes, and these differences in cellular phenotypes resulted in variance in cellular energy metabolism. Considering the enormous energy requirement of microglia for immune functions, the changes in mitochondria-centered energy metabolism and substrates of microglia are crucial for the cellular regulation of immune responses. Here we reviewed the mechanisms of microglial metabolic reprogramming by analyzing their flexible metabolic patterns and changes that occurred in their metabolism during the development of AD. Further, we summarized the role of drugs in modulating immunometabolic reprogramming to prevent neuroinflammation, which may shed light on a new research direction for AD treatment.
Collapse
Affiliation(s)
- Hongli Chen
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Zichen Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| |
Collapse
|
5
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Zhu J, Jiang X, Chang Y, Wu Y, Sun S, Wang C, Zheng S, Wang M, Yao Y, Li G, Ma R. Clemastine fumarate attenuates tauopathy and meliorates cognition in hTau mice via autophagy enhancement. Int Immunopharmacol 2023; 123:110649. [PMID: 37494840 DOI: 10.1016/j.intimp.2023.110649] [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: 04/24/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
Clemastine fumarate, which has been identified as a promising agent for remyelination and autophagy enhancement, has been shown to mitigate Aβ deposition and improve cognitive function in the APP/PS1 mouse model of Alzheimer's disease. Based on these findings, we investigated the effect of clemastine fumarate in hTau mice, a different Alzheimer's disease model characterized by overexpression of human Tau protein. Surprisingly, clemastine fumarate was effective in reducing pathological deposition of Tau protein, protecting neurons and synapses from damage, inhibiting neuroinflammation, and improving cognitive impairment in hTau mice. Interestingly, chloroquine, an autophagy inhibitor, had a significant impact on total and Sarkosyl fractions of autophagy, demonstrating that it can interrupt autophagy. Notably, after administration of chloroquine, levels of Tau protein were significantly increased. When clemastine fumarate was co-administered with chloroquine, the protective effects were reversed, indicating that clemastine fumarate indeed triggered autophagy and promoted the degradation of Tau protein, while also inhibiting further Tauopathy-related neuroinflammation and synapse loss to improve cognitive function in hTau mice.
Collapse
Affiliation(s)
- Jiahui Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Neurology, Wuhan Fourth Hospital, Wuhan 430033 Hubei, China
| | - Xingjun Jiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yanmin Chang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanqing Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shangqi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cailin Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyi Zheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Wang
- Department of Neurology, Wuhan Fourth Hospital, Wuhan 430033 Hubei, China
| | - Yi Yao
- Department of Neurology, Wuhan Fourth Hospital, Wuhan 430033 Hubei, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Jiang S, Wang X, Cao T, Kang R, Huang L. Insights on therapeutic potential of clemastine in neurological disorders. Front Mol Neurosci 2023; 16:1279985. [PMID: 37840769 PMCID: PMC10568021 DOI: 10.3389/fnmol.2023.1279985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Clemastine, a Food and Drug Administration (FDA)-approved compound, is recognized as a first-generation, widely available antihistamine that reduces histamine-induced symptoms. Evidence has confirmed that clemastine can transport across the blood-brain barrier and act on specific neurons and neuroglia to exert its protective effect. In this review, we summarize the beneficial effects of clemastine in various central nervous system (CNS) disorders, including neurodegenerative disease, neurodevelopmental deficits, brain injury, and psychiatric disorders. Additionally, we highlight key cellular links between clemastine and different CNS cells, in particular in oligodendrocyte progenitor cells (OPCs), oligodendrocytes (OLs), microglia, and neurons.
Collapse
Affiliation(s)
- Sufang Jiang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xueji Wang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tianyu Cao
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rongtian Kang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lining Huang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Ministry of Education, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
9
|
Zhu J, Ma R, Li G. Drug repurposing: Clemastine fumarate and neurodegeneration. Biomed Pharmacother 2023; 157:113904. [PMID: 36370521 DOI: 10.1016/j.biopha.2022.113904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Neurodegenerative diseases have been a weighty problem in elder people who might be stricken with motor or/and cognition defects with lower life quality urging for effective treatment. Drugs are costly from development to market, so that drug repurposing, exploration of existing drugs for novel therapeutic purposes, becomes a wise and popular strategy to raise new treatment options. Clemastine fumarate, different from anti-allergic effect as H1 histamine antagonist, was screened and identified as promising drug for remyelination and autophagy enhancement. Surprisingly, fumarate salt also has similar effect. Hence, whether clemastine fumarate would make a protective impact on neurodegenerative diseases and what contribution fumarate probably makes are intriguing to us. In this review, we summarize the potential mechanism surrounding clemastine fumarate in current literature, and try to distinguish independent or synergistic effect between clemastine and fumarate, aiming to find worthwhile research direction for neurodegeneration diseases.
Collapse
Affiliation(s)
- Jiahui Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Volonté C, Amadio S. Rethinking purinergic concepts and updating the emerging role of P2X7 and P2X4 in amyotrophic lateral sclerosis. Neuropharmacology 2022; 221:109278. [PMID: 36202258 DOI: 10.1016/j.neuropharm.2022.109278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022]
Abstract
The topic of the present review regards the ubiquitous and phylogenetically most ancient prototype of intercellular signaling, the one mediated by extracellular nucleosides and nucleotides, bearing a strong influence on pathophysiological processes in the nervous system. Not by chance, purine and pyrimidine molecules are the most prevalent and ubiquitous chemical messengers in the animal and plant kingdoms, operating through a large plethora of purinergic metabolizing enzymes, P1 and P2 receptors, nucleoside and nucleotide channels and transporters. Because ectonucleotidases degrade the agonists of P2 receptors while simultaneously generate the agonists for P1 receptors, and because several agonists, or antagonists, simultaneously bind and activate, or inhibit, more than one receptor subtype, it follows that an all-inclusive "purinergic network" perspective should be better considered when looking at purinergic actions. This becomes particularly crucial during pathological conditions as for instance amyotrophic lateral sclerosis, where the contribution of purinergic signaling has been demonstrated to differ according to each target cell phenotype and stage of disease progression. Here we will present some newly updated results about P2X7 and P2X4 as the most thoroughly investigated P2 receptors in amyotrophic lateral sclerosis, being aware that the comprehension of their actions is still in progress, and that the purinergic rationale for studying this disease must be however wide-ranging and all-inclusive. This article is part of the Special Issue on 'Purinergic Signaling: 50 years'.
Collapse
Affiliation(s)
- Cinzia Volonté
- CNR-Institute for Systems Analysis and Computer Science "Antonio Ruberti", Via Dei Taurini 19, 00185, Rome, Italy; IRCCS Fondazione Santa Lucia-Cellular Neurobiology Unit, Via Del Fosso di Fiorano 65, 00143, Rome, Italy.
| | - Susanna Amadio
- IRCCS Fondazione Santa Lucia-Cellular Neurobiology Unit, Via Del Fosso di Fiorano 65, 00143, Rome, Italy
| |
Collapse
|
11
|
Gao Y, Xie D, Wang Y, Niu L, Jiang H. Short-Chain Fatty Acids Reduce Oligodendrocyte Precursor Cells Loss by Inhibiting the Activation of Astrocytes via the SGK1/IL-6 Signalling Pathway. Neurochem Res 2022; 47:3476-3489. [PMID: 36098889 PMCID: PMC9546972 DOI: 10.1007/s11064-022-03710-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/09/2022]
Abstract
Short-chain fatty acids (SCFAs) are known to be actively involved in neurological diseases, but their roles in hypoxic-ischaemic brain injury (HIBI) are unclear. In this study, a rat model of HIBI was established, and this study measured the changes in IL-6 and NOD-like receptor thermal protein domain associated protein 3 (NLRP3), in addition to proliferation and apoptosis indicators of oligodendrocyte precursor cells (OPCs). The mechanism of action of SCFA on astrocytes was also investigated. Astrocytes were subjected to hypoxia in vitro, and OPCs were treated with IL-6. The results showed that SCFAs significantly alleviated HIBI-induced activation of astrocytes and loss of OPCs. SCFA pretreatment (1) downregulated the expression of NLRP3, IL-6, CCL2, and IP-10; (2) had no effect on the proliferation of OPCs; (3) ameliorated the abnormal expression of Bax and Bcl-2; and (4) regulated IL-6 expression via the SGK1-related pathway in astrocytes. Our findings revealed that SCFAs alleviated the loss of OPCs by regulating astrocyte activation through the SGK1/IL-6 signalling pathway.
Collapse
Affiliation(s)
- Yanmin Gao
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Department of General Practice, Kongjiang Community Health Service Center, No. 100, Yanji West Road, Yangpu District, Shanghai, 200093, China
| | - Di Xie
- Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yang Wang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Lei Niu
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Hua Jiang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China. .,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
12
|
Abdelhak A, Cordano C, Boscardin WJ, Caverzasi E, Kuhle J, Chan B, Gelfand JM, Yiu HH, Oertel FC, Beaudry-Richard A, Condor Montes S, Oksenberg JR, Lario Lago A, Boxer A, Rojas-Martinez JC, Elahi FM, Chan JR, Green AJ. Plasma neurofilament light chain levels suggest neuroaxonal stability following therapeutic remyelination in people with multiple sclerosis. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2022-329221. [PMID: 35710320 PMCID: PMC9984688 DOI: 10.1136/jnnp-2022-329221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/23/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Chronic demyelination is a major contributor to axonal vulnerability in multiple sclerosis (MS). Therefore, remyelination could provide a potent neuroprotective strategy. The ReBUILD trial was the first study showing evidence for successful remyelination following treatment with clemastine in people with MS (pwMS) with no evidence of disease activity or progression (NEDAP). Whether remyelination was associated with neuroprotection remains unexplored. METHODS Plasma neurofilament light chain (NfL) levels were measured from ReBUILD trial's participants. Mixed linear effect models were fit for individual patients, epoch and longitudinal measurements to compare NfL concentrations between samples collected during the active and placebo treatment period. RESULTS NfL concentrations were 9.6% lower in samples collected during the active treatment with clemastine (n=53, geometric mean=6.33 pg/mL) compared to samples collected during treatment with placebo (n=73, 7.00 pg/mL) (B=-0.035 [-0.068 to -0.001], p=0.041). Applying age- and body mass index-standardised NfL Z-scores and percentiles revealed similar results (0.04 vs 0.35, and 27.5 vs 33.3, p=0.023 and 0.042, respectively). Higher NfL concentrations were associated with more delayed P100 latencies (B=1.33 [0.26 to 2.41], p=0.015). In addition, improvement of P100 latencies between visits was associated with a trend for lower NfL values (B=0.003 [-0.0004 to 0.007], p=0.081). Based on a Cohen's d of 0.248, a future 1:1 parallel-arm placebo-controlled study using a remyelinating agent with comparable effect as clemastine would need 202 subjects per group to achieve 80% power. CONCLUSIONS In pwMS, treatment with the remyelinating agent clemastine was associated with a reduction of blood NfL, suggesting that neuroprotection is achievable and measurable with therapeutic remyelination. TRIAL REGISTRATION NUMBER NCT02040298.
Collapse
Affiliation(s)
- Ahmed Abdelhak
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - W John Boscardin
- Departments of Medicine and Epidemiology & Biostatistics, University of California at San Francisco, San Francisco, California, USA
| | - Eduardo Caverzasi
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Jens Kuhle
- Multiple Sclerosis Centre, Neurology, Departments of Head, Spine and Neuromedicine, Biomedicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Brandon Chan
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Jeffrey M Gelfand
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Hao H Yiu
- Department of Biology, University of Maryland, College Park, Maryland, USA
| | - Frederike C Oertel
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Alexandra Beaudry-Richard
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Shivany Condor Montes
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Argentina Lario Lago
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Adam Boxer
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Julio C Rojas-Martinez
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Fanny M Elahi
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Jonah R Chan
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| | - Ari J Green
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco (UCSF), San Francisco, California, USA
| |
Collapse
|
13
|
Begum R, Thota S, Abdulkadir A, Kaur G, Bagam P, Batra S. NADPH oxidase family proteins: signaling dynamics to disease management. Cell Mol Immunol 2022; 19:660-686. [PMID: 35585127 DOI: 10.1038/s41423-022-00858-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 03/12/2022] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) are pervasive signaling molecules in biological systems. In humans, a lack of ROS causes chronic and extreme bacterial infections, while uncontrolled release of these factors causes pathologies due to excessive inflammation. Professional phagocytes such as neutrophils (PMNs), eosinophils, monocytes, and macrophages use superoxide-generating NADPH oxidase (NOX) as part of their arsenal of antimicrobial mechanisms to produce high levels of ROS. NOX is a multisubunit enzyme complex composed of five essential subunits, two of which are localized in the membrane, while three are localized in the cytosol. In resting phagocytes, the oxidase complex is unassembled and inactive; however, it becomes activated after cytosolic components translocate to the membrane and are assembled into a functional oxidase. The NOX isoforms play a variety of roles in cellular differentiation, development, proliferation, apoptosis, cytoskeletal control, migration, and contraction. Recent studies have identified NOX as a major contributor to disease pathologies, resulting in a shift in focus on inhibiting the formation of potentially harmful free radicals. Therefore, a better understanding of the molecular mechanisms and the transduction pathways involved in NOX-mediated signaling is essential for the development of new therapeutic agents that minimize the hyperproduction of ROS. The current review provides a thorough overview of the various NOX enzymes and their roles in disease pathophysiology, highlights pharmacological strategies, and discusses the importance of computational modeling for future NOX-related studies.
Collapse
Affiliation(s)
- Rizwana Begum
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Shilpa Thota
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Abubakar Abdulkadir
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Gagandeep Kaur
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA.,Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA.,Division of Systems Biology, National Center for Toxicological Research, Jefferson, AR, 72079, USA
| | - Sanjay Batra
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
14
|
Bassani D, Pavan M, Federico S, Spalluto G, Sturlese M, Moro S. The Multifaceted Role of GPCRs in Amyotrophic Lateral Sclerosis: A New Therapeutic Perspective? Int J Mol Sci 2022; 23:4504. [PMID: 35562894 PMCID: PMC9106011 DOI: 10.3390/ijms23094504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/15/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerating disease involving the motor neurons, which causes a progressive loss of movement ability, usually leading to death within 2 to 5 years from the diagnosis. Much effort has been put into research for an effective therapy for its eradication, but still, no cure is available. The only two drugs approved for this pathology, Riluzole and Edaravone, are onlyable to slow down the inevitable disease progression. As assessed in the literature, drug targets such as protein kinases have already been extensively examined as potential drug targets for ALS, with some molecules already in clinical trials. Here, we focus on the involvement of another very important and studied class of biological entities, G protein-coupled receptors (GPCRs), in the onset and progression of ALS. This workaimsto give an overview of what has been already discovered on the topic, providing useful information and insights that can be used by scientists all around the world who are putting efforts into the fight against this very important neurodegenerating disease.
Collapse
Affiliation(s)
- Davide Bassani
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| |
Collapse
|
15
|
Guo S, Wang H, Yin Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:815347. [PMID: 35250543 PMCID: PMC8888930 DOI: 10.3389/fnagi.2022.815347] [Citation(s) in RCA: 320] [Impact Index Per Article: 106.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia-mediated neuroinflammation is a common feature of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Microglia can be categorized into two opposite types: classical (M1) or alternative (M2), though there’s a continuum of different intermediate phenotypes between M1 and M2, and microglia can transit from one phenotype to another. M1 microglia release inflammatory mediators and induce inflammation and neurotoxicity, while M2 microglia release anti-inflammatory mediators and induce anti-inflammatory and neuroprotectivity. Microglia-mediated neuroinflammation is considered as a double-edged sword, performing both harmful and helpful effects in neurodegenerative diseases. Previous studies showed that balancing microglia M1/M2 polarization had a promising therapeutic prospect in neurodegenerative diseases. We suggest that shifting microglia from M1 to M2 may be significant and we focus on the modulation of microglia polarization from M1 to M2, especially by important signal pathways, in neurodegenerative diseases.
Collapse
|
16
|
Chen JF, Wang F, Huang NX, Xiao L, Mei F. Oligodendrocytes and Myelin: Active players in Neurodegenerative brains? Dev Neurobiol 2022; 82:160-174. [PMID: 35081276 DOI: 10.1002/dneu.22867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/10/2022]
Abstract
Oligodendrocytes (OLs) are a major type of glial cells in the central nervous system that generate multiple myelin sheaths to wrap axons. Myelin ensures fast and efficient propagation of action potentials along axons and supports neurons with nourishment. The decay of OLs and myelin has been implicated in age-related neurodegenerative diseases and these changes are generally considered as an inevitable result of neuron loss and axon degeneration. Noticeably, OLs and myelin undergo dynamic changes in healthy adult brains, that is, newly formed OLs are continuously added throughout life from the differentiation of oligodendrocyte precursor cells (OPCs) and the pre-existing myelin sheaths may undergo degeneration or remodeling. Increasing evidence has shown that changes in OLs and myelin are present in the early stages of neurodegenerative diseases, and even prior to significant neuronal loss and functional deficits. More importantly, oligodendroglia-specific manipulation, by either deletion of the disease gene or enhancement of myelin renewal, can alleviate functional impairments in neurodegenerative animal models. These findings underscore the possibility that OLs and myelin are not passively but actively involved in neurodegenerative diseases and may play an important role in modulating neuronal function and survival. In this review, we summarize recent work characterizing OL and myelin changes in both healthy and neurodegenerative brains and discuss the potential of targeting oligodendroglial cells in treating neurodegenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Nan-Xing Huang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
17
|
Clemastine Rescues Chemotherapy-Induced Cognitive Impairment by Improving White Matter Integrity. Neuroscience 2022; 484:66-79. [PMID: 35007691 DOI: 10.1016/j.neuroscience.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
With the improvement of cancer treatment techniques, increasing attention has been given to chemotherapy-induced cognitive impairment through white matter injury. Clemastine fumarate has been shown to enhance white matter integrity in cuprizone- or hypoxia-induced demyelination mouse models. However, whether clemastine can be beneficial for reversing chemotherapy-induced cognitive impairment remains unexplored. In this study, the mice received oral administration of clemastine after chemotherapy. The open-field test and Morris water maze test were used to evaluate their anxiety, locomotor activity and cognitive function. Luxol Fast Blue staining and transmission electron microscopy were used to detect the morphological damage to the myelin. Demyelination and damage to the mature oligodendrocytes and axons were observed by immunofluorescence and western blotting. Clemastine significantly improved their cognitive function and ameliorated white matter injury in the chemotherapy-treated mice. Clemastine enhanced myelination, promoted oligodendrocyte precursor cell differentiation and increased the neurofilament 200 protein levels in the corpus callosum and hippocampus. We concluded that clemastine rescues cognitive function damage caused by chemotherapy through improving white matter integrity. Remyelination, oligodendrocyte differentiation and the increase of neurofilament protein promoted by clemastine are potential strategies for reversing the cognitive dysfunction caused by chemotherapy.
Collapse
|
18
|
Abstract
Microglia, a category of glial cells in the central nervous system (CNS), have attracted much attention because of their important role in neuroinflammation. Many translational studies are currently ongoing to discover novel drugs targeting microglia for the treatment of various CNS disorders, such as Alzheimer's disease, Parkinson's disease (PD), and depression. Recent studies have shown that brain histamine, a neurotransmitter essential for the regulation of diverse brain functions, controls glial cells and neurons. In vitro studies using primary microglia and microglial cell lines have reported that histamine receptors are expressed in microglia and control microglial functions, including chemotaxis, migration, cytokine secretion, and autophagy. In vivo studies have demonstrated that histamine-related reagents could ameliorate abnormal symptoms in animal models of human diseases, such as amyotrophic lateral sclerosis (ALS), PD, and brain ischemia. Several human studies have revealed alterations in histamine receptor levels in ALS and PD, emphasizing the importance of the CNS histamine system, including histamine-dependent microglial modulation, as a therapeutic target for these disorders. In this review article, we summarize histamine-related research focusing on microglial functions.
Collapse
Affiliation(s)
- Tomomitsu Iida
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
19
|
Polaryzacja mikrogleju i makrofagów w wybranych chorobach degeneracyjnych i zapalnych układu nerwowego. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Makrofagi to komórki efektorowe układu odpornościowego zdolne do polaryzacji, czyli zmiany fenotypu powiązanej ze zmianą aktywności. Można wyróżnić: polaryzację klasyczną (M1), która służy obronie przed patogenami, a makrofagi M1 mają aktywność ogólnie prozapalną, oraz polaryzację alternatywną (M2), która sprzyja wygaszaniu stanu zapalnego i regeneracji tkanki. Makrofagi zasiedlają niemal cały organizm, więc zjawisko ich polaryzacji ma wpływ na wiele procesów zachodzących w różnych tkankach. W układzie nerwowym reprezentacją osiadłych makrofagów jest mikroglej. Jednak w wielu sytuacjach patologicznych w mózgu pojawiają się także makrofagi rekrutowane z monocytów krążących we krwi. Choroby neurodegeneracyjne, urazy i choroby autoimmunologiczne są związane z reakcją układu odpornościowego, która może mieć istotny wpływ na dalszy przebieg choroby i na tempo regeneracji tkanki. Polaryzacja makrofagów ma w związku z tym znaczenie w chorobach centralnego układu nerwowego. Aktywność komórek M1 i M2 może bowiem różnie wpływać na przeżywalność neuronów i oligodendrocytów, na wzrost aksonów, na proces demielinizacji czy na szczelność bariery krew–mózg. Wynika to z różnic między fenotypami w wytwarzaniu reaktywnych form tlenu i tlenku azotu, wydzielaniu cytokin i czynników wzrostu, bezpośrednich oddziaływaniach na sąsiednie komórki i zdolnościach do fagocytozy. W artykule omówiono to zagadnienie w: udarze mózgu, urazie rdzenia kręgowego, chorobie Alzheimera, stwardnieniu zanikowym bocznym i stwardnieniu rozsianym. W wielu spośród tych patologii obserwuje się gradient czasowy lub przestrzenny rozmieszczenia w tkance poszczególnych fenotypów mikrogleju i/lub makrofagów. Wydaje się zatem, że zmiany polaryzacji makrofagów mogą potencjalnie sprzyjać regeneracji tkanki lub hamować rozwój chorób neurodegeneracyjnych.
Collapse
|
20
|
Benarroch E. What Is the Role of Oligodendrocytes in Amyotrophic Lateral Sclerosis? Neurology 2021; 97:776-779. [PMID: 34663738 DOI: 10.1212/wnl.0000000000012706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 11/15/2022] Open
|
21
|
Novel P2X7 Antagonist Ameliorates the Early Phase of ALS Disease and Decreases Inflammation and Autophagy in SOD1-G93A Mouse Model. Int J Mol Sci 2021; 22:ijms221910649. [PMID: 34638992 PMCID: PMC8508678 DOI: 10.3390/ijms221910649] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease with a resilient neuroinflammatory component caused by activated microglia and infiltrated immune cells. How to successfully balance neuroprotective versus neurotoxic actions through the use of anti-inflammatory agents is still under debate. There has been a boost of awareness regarding the role of extracellular ATP and purinergic receptors in modulating the physiological and pathological mechanisms in the nervous system. Particularly in ALS, it is known that the purinergic ionotropic P2X7 receptor plays a dual role in disease progression by acting at different cellular and molecular levels. In this context, we previously demonstrated that the P2X7 receptor antagonist, brilliant blue G, reduces neuroinflammation and ameliorates some of the pathological features of ALS in the SOD1-G93A mouse model. Here, we test the novel, noncommercially available, and centrally permeant Axxam proprietary P2X7 antagonist, AXX71, in SOD1-G93A mice, by assessing some behavioral and molecular parameters, among which are disease progression, survival, gliosis, and motor neuron wealth. We demonstrate that AXX71 affects the early symptomatic phase of the disease by reducing microglia-related proinflammatory markers and autophagy without affecting the anti-inflammatory markers or motor neuron survival. Our results suggest that P2X7 modulation can be further investigated as a therapeutic strategy in preclinical studies, and exploited in ALS clinical trials.
Collapse
|
22
|
Fiscon G, Conte F, Amadio S, Volonté C, Paci P. Drug Repurposing: A Network-based Approach to Amyotrophic Lateral Sclerosis. Neurotherapeutics 2021; 18:1678-1691. [PMID: 33987813 PMCID: PMC8609089 DOI: 10.1007/s13311-021-01064-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
The continuous adherence to the conventional "one target, one drug" paradigm has failed so far to provide effective therapeutic solutions for heterogeneous and multifactorial diseases as amyotrophic lateral sclerosis (ALS), a rare progressive and chronic, debilitating neurological disease for which no cure is available. The present study is aimed at finding innovative solutions and paradigms for therapy in ALS pathogenesis, by exploiting new insights from Network Medicine and drug repurposing strategies. To identify new drug-ALS disease associations, we exploited SAveRUNNER, a recently developed network-based algorithm for drug repurposing, which quantifies the proximity of disease-associated genes to drug targets in the human interactome. We prioritized 403 SAveRUNNER-predicted drugs according to decreasing values of network similarity with ALS. Among catecholamine, dopamine, serotonin, histamine, and GABA receptor modulators, as well as angiotensin-converting enzymes, cyclooxygenase isozymes, and serotonin transporter inhibitors, we found some interesting no customary ALS drugs, including amoxapine, clomipramine, mianserin, and modafinil. Furthermore, we strengthened the SAveRUNNER predictions by a gene set enrichment analysis that confirmed modafinil as a drug with the highest score among the 121 identified drugs with a score > 0. Our results contribute to gathering further proofs of innovative solutions for therapy in ALS pathogenesis.
Collapse
Affiliation(s)
- Giulia Fiscon
- Institute for Systems Analysis and Computer Science “A. Ruberti”, National Research Council (IASI–CNR), Via Dei Taurini 19, 00185 Rome, Italy
- Fondazione per la Medicina Personalizzata, Via Goffredo Mameli, Genova, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science “A. Ruberti”, National Research Council (IASI–CNR), Via Dei Taurini 19, 00185 Rome, Italy
| | - Susanna Amadio
- IRCCS Santa Lucia Foundation, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Cinzia Volonté
- Institute for Systems Analysis and Computer Science “A. Ruberti”, National Research Council (IASI–CNR), Via Dei Taurini 19, 00185 Rome, Italy
- IRCCS Santa Lucia Foundation, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Paola Paci
- Institute for Systems Analysis and Computer Science “A. Ruberti”, National Research Council (IASI–CNR), Via Dei Taurini 19, 00185 Rome, Italy
- Department of Computer, Control, and Management Engineering Antonio Ruberti (DIAG), Sapienza University, Rome, Italy
| |
Collapse
|
23
|
Clemastine attenuates AD-like pathology in an AD model mouse via enhancing mTOR-mediated autophagy. Exp Neurol 2021; 342:113742. [PMID: 33965410 DOI: 10.1016/j.expneurol.2021.113742] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 04/07/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with limited available drugs for treatment. Enhancing autophagy attenuates AD pathology in various AD model mice. Thus, development of potential drugs which enhance autophagy may bring beneficial effects in AD therapy. In the present study, we show clemastine, a first-generation histamine H1R antagonist and being originally marketed for the treatment of allergic rhinitis, ameliorates AD pathogenesis in APP/PS1 transgenic mice. Chronic treatment with clemastine orally reduced amyloid-β (Aβ) load, neuroinflammation and cognitive deficits of APP/PS1 transgenic mice. Clemastine decreases Aβ generation via reducing the levels of BACE1, CTFs of APP. Mechanistically, clemastine enhances autophagy concomitant with a suppression of mTOR signaling. Therefore, we propose that clemastine attenuates AD pathology via enhancing mTOR-mediated autophagy.
Collapse
|
24
|
Chenchula S, Ray A, Sadasivam B. Famotidine Repurposing for Novel Corona Virus Disease of 2019: A
Systematic Review. Drug Res (Stuttg) 2021; 71:295-301. [DOI: 10.1055/a-1397-6763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Abstract
Background COVID-19 caused by SARS-CoV-2 was declared as a global
pandemic by the WHO. Famotidine is a histamine-2 (H2) receptor antagonist
which blocks the H2 receptors in the parietal cells, decreasing gastric acid
secretion. Our review aims to study all the available scientific evidence on
famotidine research outcomes systematically to introspect its clinical
efficacy and probable mechanisms and clinical efficacy against
SARS-CoV-2.
Methodology An electronic search of PubMed, Scopus and Google Scholar
was performed using MeSH terms “SARS CoV-2” OR
“COVID-19” AND“FAMOTIDINE”. Relevant
informationwas extracted from studies reporting the efficacy of famotidine
in COVID-19.
Results We found a total of 32 studies, out of which only 14 were
relevant and were included in our review.Molecular computational studies
showed that famotidine selectively acts on viral replication proteases
papain-like protease (PLpro) and 3-chymotrypsin-like protease (3CLpro).
Additionally, it acts via inverse-agonism on the H2 receptors present in
neutrophils and eosinophils which leads to inhibition of cytokine release.
Clinical study findings have pointed toward significant improvements in
COVID-19 patient-reported symptoms in non-hospitalized patients and
reduction in intubation or death in critically ill patients associated with
the usage of famotidine. However,in one of the studies,famotidine has failed
to show any significant benefit in reducing mortality due to COVID-19.
Conclusion Famotidine has the potential to answer the ongoing global
challenge owing to its selective action on viral replication. Additionally,
clinical findings in COVID-19 patients support its efficacy to reduce
clinical symptoms of COVID-19.We suggest that further optimally powered
randomized clinical trials should be carried out to come up with definitive
conclusions.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences
Bhopal, Bhopal, Madhya Pradesh, India
| | - Avik Ray
- Department of Pharmacology, All India Institute of Medical Sciences
Bhopal, Bhopal, Madhya Pradesh, India
| | - Balakrishnan Sadasivam
- Department of Pharmacology, All India Institute of Medical Sciences
Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
25
|
Raffaele S, Boccazzi M, Fumagalli M. Oligodendrocyte Dysfunction in Amyotrophic Lateral Sclerosis: Mechanisms and Therapeutic Perspectives. Cells 2021; 10:cells10030565. [PMID: 33807572 PMCID: PMC8000560 DOI: 10.3390/cells10030565] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Myelin is the lipid-rich structure formed by oligodendrocytes (OLs) that wraps the axons in multilayered sheaths, assuring protection, efficient saltatory signal conduction and metabolic support to neurons. In the last few years, the impact of OL dysfunction and myelin damage has progressively received more attention and is now considered to be a major contributing factor to neurodegeneration in several neurological diseases, including amyotrophic lateral sclerosis (ALS). Upon OL injury, oligodendrocyte precursor cells (OPCs) of adult nervous tissue sustain the generation of new OLs for myelin reconstitution, but this spontaneous regeneration process fails to successfully counteract myelin damage. Of note, the functions of OPCs exceed the formation and repair of myelin, and also involve the trophic support to axons and the capability to exert an immunomodulatory role, which are particularly relevant in the context of neurodegeneration. In this review, we deeply analyze the impact of dysfunctional OLs in ALS pathogenesis. The possible mechanisms underlying OL degeneration, defective OPC maturation, and impairment in energy supply to motor neurons (MNs) have also been examined to provide insights on future therapeutic interventions. On this basis, we discuss the potential therapeutic utility in ALS of several molecules, based on their remyelinating potential or capability to enhance energy metabolism.
Collapse
|
26
|
Deng H, Zhang Y, Li GG, Yu HH, Bai S, Guo GY, Guo WL, Ma Y, Wang JH, Liu N, Pan C, Tang ZP. P2X7 receptor activation aggravates NADPH oxidase 2-induced oxidative stress after intracerebral hemorrhage. Neural Regen Res 2021; 16:1582-1591. [PMID: 33433488 PMCID: PMC8323669 DOI: 10.4103/1673-5374.303036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress is a crucial pathological process that contributes to secondary injury following intracerebral hemorrhage. P2X7 receptor (P2X7R), which is activated by the abnormal accumulation of extracellular ATP, plays an important role in the regulation of oxidative stress in the central nervous system, although the effects of activated P2X7R-associated oxidative stress after intracerebral hemorrhage remain unclear. Mouse models of intracerebral hemorrhage were established through the stereotactic injection of 0.075 U VII collagenase into the right basal ganglia. The results revealed that P2X7R expression peaked 24 hours after intracerebral hemorrhage, and P2X7R expressed primarily in neurons. The inhibition of P2X7R, using A438079 (100 mg/kg, intraperitoneal), reduced nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) expression and malondialdehyde generation, increased superoxide dismutase and glutathione/oxidized glutathione levels, and alleviated neurological damage, brain edema, and apoptosis after intracellular hemorrhage. The P2X7R inhibitor A438079 (100 mg/kg, intraperitoneal injection) inhibited the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and nuclear factor kappa-B (NF-κB) after intracerebral hemorrhage. Blocking ERK1/2 activation, using the ERK1/2 inhibitor U0126 (2 µg, intraventricular injection), reduced the level of NOX2-mediated oxidative stress induced by P2X7R activation after intracellular hemorrhage. Similarly, the inhibition of NF-κB, using the NF-κB inhibitor JSH-23 (3.5 µg, intraventricular), reduced the level of NOX2-mediated oxidative stress induced by P2X7R activation. Finally, GSK2795039 (100 mg/kg, intraperitoneal), a NOX2 antagonist, attenuated P2X7R-mediated oxidative stress, neurological damage, and brain edema after intracerebral hemorrhage. The results indicated that P2X7R activation aggravated NOX2-induced oxidative stress through the activation of the ERK1/2 and NF-κB pathways following intracerebral hemorrhage in mice. The present study was approved by the Ethics Committee of Huazhong University of Science and Technology, China (approval No. TJ-A20160805) on August 26, 2016.
Collapse
Affiliation(s)
- Hong Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ye Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Gai-Gai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hai-Han Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuang Bai
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Guang-Yu Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wen-Liang Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yang Ma
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Hui Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
27
|
Angelini DF, De Angelis F, Vacca V, Piras E, Parisi C, Nutini M, Spalloni A, Pagano F, Longone P, Battistini L, Pavone F, Marinelli S. Very Early Involvement of Innate Immunity in Peripheral Nerve Degeneration in SOD1-G93A Mice. Front Immunol 2020; 11:575792. [PMID: 33329541 PMCID: PMC7714949 DOI: 10.3389/fimmu.2020.575792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Recent preclinical and clinical evidence suggest that immune system has a role in the progression and prognosis of Amyotrophic Lateral Sclerosis (ALS), but the identification of a clear mechanism and immune players remains to be elucidated. Here, we have investigated, in 30 and 60 days (presymptomatic) and 120 days (symptomatic) old SOD1-G93A mice, systemic, peripheral, and central innate and adaptive immune and inflammatory response, correlating it with the progression of the neurodegeneration in neuromuscular junction, sciatic nerves, and spinal cord. Surprisingly, we found a very initial (45-60 days) presence of IgG in sciatic nerves together with a gradual enhancement of A20/TNFAIP3 (protein controlling NF-κB signalling) and a concomitantly significant increase and activation of circulating mast cells (MCs) as well as MCs and macrophages in sciatic nerve and an enhancement of IL-6 and IL-10. This immunological frame coincided with a myelin aggregation. The 30-60 days old SOD1-G93A mice didn't show real elements of neuroinflammation and neurodegeneration in spinal cord. In 120 days old mice macrophages and monocytes are widely diffused in sciatic nerves, peripheral neurodegeneration reaches the tip, high circulating levels of TNFα and IL-2 were found and spinal cord exhibits clear signs of neural damage and infiltrating immune cells. Our results underpin a clear immunological disorder at the origin of ALS axonopathy, in which MCs are involved in the initiation and sustaining of inflammatory events. These data cannot be considered a mere epiphenomenon of motor neuron degeneration and reveal new potential selective immune targets in ALS therapy.
Collapse
Affiliation(s)
| | - Federica De Angelis
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Valentina Vacca
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Eleonora Piras
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Chiara Parisi
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Michele Nutini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Alida Spalloni
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesca Pagano
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Flaminia Pavone
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Sara Marinelli
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| |
Collapse
|
28
|
Amin A, Perera ND, Beart PM, Turner BJ, Shabanpoor F. Amyotrophic Lateral Sclerosis and Autophagy: Dysfunction and Therapeutic Targeting. Cells 2020; 9:E2413. [PMID: 33158177 PMCID: PMC7694295 DOI: 10.3390/cells9112413] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past 20 years, there has been a drastically increased understanding of the genetic basis of Amyotrophic Lateral Sclerosis. Despite the identification of more than 40 different ALS-causing mutations, the accumulation of neurotoxic misfolded proteins, inclusions, and aggregates within motor neurons is the main pathological hallmark in all cases of ALS. These protein aggregates are proposed to disrupt cellular processes and ultimately result in neurodegeneration. One of the main reasons implicated in the accumulation of protein aggregates may be defective autophagy, a highly conserved intracellular "clearance" system delivering misfolded proteins, aggregates, and damaged organelles to lysosomes for degradation. Autophagy is one of the primary stress response mechanisms activated in highly sensitive and specialised neurons following insult to ensure their survival. The upregulation of autophagy through pharmacological autophagy-inducing agents has largely been shown to reduce intracellular protein aggregate levels and disease phenotypes in different in vitro and in vivo models of neurodegenerative diseases. In this review, we explore the intriguing interface between ALS and autophagy, provide a most comprehensive summary of autophagy-targeted drugs that have been examined or are being developed as potential treatments for ALS to date, and discuss potential therapeutic strategies for targeting autophagy in ALS.
Collapse
Affiliation(s)
| | | | | | | | - Fazel Shabanpoor
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia; (A.A.); (N.D.P.); (P.M.B.); (B.J.T.)
| |
Collapse
|
29
|
Volonté C, Morello G, Spampinato AG, Amadio S, Apolloni S, D’Agata V, Cavallaro S. Omics-based exploration and functional validation of neurotrophic factors and histamine as therapeutic targets in ALS. Ageing Res Rev 2020; 62:101121. [PMID: 32653439 DOI: 10.1016/j.arr.2020.101121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/27/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
A plethora of genetic and molecular mechanisms have been implicated in the pathophysiology of the heterogeneous and multifactorial amyotrophic lateral sclerosis (ALS) disease, and hence the conventional "one target-one drug" paradigm has failed so far to provide effective therapeutic solutions, precisely because of the complex nature of ALS. This review intends to highlight how the integration of emerging "omics" approaches may provide a rational foundation for the comprehensive exploration of molecular pathways and dynamic interactions involved in ALS, for the identification of candidate targets and biomarkers that will assist in the rapid diagnosis and prognosis, lastly for the stratification of patients into different subgroups with the aim of personalized therapeutic strategies. To this purpose, particular emphasis will be placed on some potential therapeutic targets, including neurotrophic factors and histamine signaling that both have emerged as dysregulated at different omics levels in specific subgroups of ALS patients, and have already shown promising results in in vitro and in vivo models of ALS. To conclude, we will discuss about the utility of using integrated omics coupled with network-based approaches to provide additional guidance for personalization of medicine applications in ALS.
Collapse
|
30
|
Wang Y, Patani R. Novel therapeutic targets for amyotrophic lateral sclerosis: ribonucleoproteins and cellular autonomy. Expert Opin Ther Targets 2020; 24:971-984. [PMID: 32746659 DOI: 10.1080/14728222.2020.1805734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a devastating disease with a lifetime risk of approximately 1:400. It is incurable and invariably fatal. Average survival is between 3 and 5 years and patients become increasingly paralyzed, losing the ability to speak, eat, and breathe. Therapies in development either (i) target specific familial forms of ALS (comprising a minority of around 10% of cases) or ii) emanate from (over)reliance on animal models or non-human/non-neuronal cell models. There is a desperate and unmet clinical need for effective treatments. Deciphering the primacy and relative contributions of defective protein homeostasis and RNA metabolism in ALS across different model systems will facilitate the identification of putative therapeutic targets. AREAS COVERED This review examines the putative common primary molecular events that lead to ALS pathogenesis. We focus on deregulated RNA metabolism, protein mislocalization/pathological aggregation and the role of glia in ALS-related motor neuron degeneration. Finally, we describe promising targets for therapeutic evaluation. EXPERT OPINION Moving forward, an effective strategy could be achieved by a poly-therapeutic approach which targets both deregulated RNA metabolism and protein dyshomeostasis in the relevant cell types, at the appropriate phase of disease.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London , London, UK.,Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute , London, UK
| | - Rickie Patani
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London , London, UK.,Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute , London, UK
| |
Collapse
|
31
|
Kataura T, Tashiro E, Nishikawa S, Shibahara K, Muraoka Y, Miura M, Sakai S, Katoh N, Totsuka M, Onodera M, Shin-Ya K, Miyamoto K, Sasazawa Y, Hattori N, Saiki S, Imoto M. A chemical genomics-aggrephagy integrated method studying functional analysis of autophagy inducers. Autophagy 2020; 17:1856-1872. [PMID: 32762399 PMCID: PMC8386610 DOI: 10.1080/15548627.2020.1794590] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macroautophagy/autophagy plays a critical role in the pathogenesis of various human diseases including neurodegenerative disorders such as Parkinson disease (PD) and Huntington disease (HD). Chemical autophagy inducers are expected to serve as disease-modifying agents by eliminating cytotoxic/damaged proteins. Although many autophagy inducers have been identified, their precise molecular mechanisms are not fully understood because of the complicated crosstalk among signaling pathways. To address this issue, we performed several chemical genomic analyses enabling us to comprehend the dominancy among the autophagy-associated pathways followed by an aggresome-clearance assay. In a first step, more than 400 target-established small molecules were assessed for their ability to activate autophagic flux in neuronal PC12D cells, and we identified 39 compounds as autophagy inducers. We then profiled the autophagy inducers by testing their effect on the induction of autophagy by 200 well-established signal transduction modulators. Our principal component analysis (PCA) and clustering analysis using a dataset of "autophagy profiles" revealed that two Food and Drug Administration (FDA)-approved drugs, memantine and clemastine, activate endoplasmic reticulum (ER) stress responses, which could lead to autophagy induction. We also confirmed that SMK-17, a recently identified autophagy inducer, induced autophagy via the PRKC/PKC-TFEB pathway, as had been predicted from PCA. Finally, we showed that almost all of the autophagy inducers tested in this present work significantly enhanced the clearance of the protein aggregates observed in cellular models of PD and HD. These results, with the combined approach, suggested that autophagy-activating small molecules may improve proteinopathies by eliminating nonfunctional protein aggregates.Abbreviations: ADK: adenosine kinase; AMPK: AMP-activated protein kinase; ATF4: activating transcription factor 4; BECN1: beclin-1; DDIT3/CHOP: DNA damage inducible transcript 3; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; EIF2S1/eIF2α: eukaryotic translation initiation factor 2 subunit alpha; ER: endoplasmic reticulum; ERN1/IRE1α: endoplasmic reticulum to nucleus signaling 1; FDA: Food and Drug Administration; GSH: glutathione; HD: Huntington disease; HSPA5/GRP78: heat shock protein family A (Hsp70) member 5; HTT: huntingtin; JAK: Janus kinase, MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MAP2K/MEK: mitogen-activated protein kinase kinase; MAP3K8/Tpl2: mitogen-activated protein kinase kinase kinase 8; MAPK: mitogen-activated protein kinase; MPP+: 1-methyl-4-phenylpyridinium; MTOR: mechanistic target of rapamycin kinase; MTORC: MTOR complex; NAC: N-acetylcysteine; NGF: nerve growth factor 2; NMDA: N-methyl-D-aspartate; PCA: principal component analysis; PD: Parkinson disease; PDA: pancreatic ductal adenocarcinoma; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PMA: phorbol 12-myristate 13-acetate; PRKC/PKC: protein kinase C; ROCK: Rho-associated coiled-coil protein kinase; RR: ribonucleotide reductase; SIGMAR1: sigma non-opioid intracellular receptor 1; SQSTM1/p62: sequestosome 1; STK11/LKB1: serine/threonine kinase 11; TFEB: Transcription factor EB; TGFB/TGF-β: Transforming growth factor beta; ULK1: unc-51 like autophagy activating kinase 1; XBP1: X-box binding protein 1.
Collapse
Affiliation(s)
- Tetsushi Kataura
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan.,Research Fellow of the Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Etsu Tashiro
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Shota Nishikawa
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Kensuke Shibahara
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Yoshihito Muraoka
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Masahiro Miura
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Shun Sakai
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Naohiro Katoh
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Misato Totsuka
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| | - Masafumi Onodera
- Division of Immunology, National Center for Child Health and Development, Tokyo, Japan
| | - Kazuo Shin-Ya
- National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,Biotechnology Research Centre, The University of Tokyo, Tokyo, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, Japan
| | - Kengo Miyamoto
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yukiko Sasazawa
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan.,Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Masaya Imoto
- Department of Biosciences and Informatics, Keio University, Kanagawa, Japan
| |
Collapse
|
32
|
Ashford BA, Boche D, Cooper-Knock J, Heath PR, Simpson JE, Highley JR. Review: Microglia in motor neuron disease. Neuropathol Appl Neurobiol 2020; 47:179-197. [PMID: 32594542 DOI: 10.1111/nan.12640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023]
Abstract
Motor Neuron Disease (MND) is a fatal neurodegenerative condition, which is characterized by the selective loss of the upper and lower motor neurons. At the sites of motor neuron injury, accumulation of activated microglia, the primary immune cells of the central nervous system, is commonly observed in both human post mortem studies and animal models of MND. Microglial activation has been found to correlate with many clinical features and importantly, the speed of disease progression in humans. Both anti-inflammatory and pro-inflammatory microglial responses have been shown to influence disease progression in humans and models of MND. As such, microglia could both contribute to and protect against inflammatory mechanisms of pathogenesis in MND. While murine models have characterized the microglial response to MND, these studies have painted a complex and often contradictory picture, indicating a need for further characterization in humans. This review examines the potential role microglia play in MND in human and animal studies. Both the pro-inflammatory and anti-inflammatory responses will be addressed, throughout the course of disease, followed by the potential of microglia as a target in the development of disease-modifying treatments for MND.
Collapse
Affiliation(s)
| | - D Boche
- University of Southampton, Southampton, UK
| | | | - P R Heath
- University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
33
|
Rodrigues AM, Serralha RS, Lima DY, Punaro GR, Visona I, Fernandes MJS, Higa EMS. P2X7 siRNA targeted to the kidneys increases klotho and delays the progression of experimental diabetic nephropathy. Purinergic Signal 2020; 16:175-185. [PMID: 32377919 PMCID: PMC7367965 DOI: 10.1007/s11302-020-09695-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/06/2020] [Indexed: 01/22/2023] Open
Abstract
Previous studies in our laboratory have suggested that P2X7 could contribute to the progression of diabetic nephropathy and modulated klotho expression. The aim of this study was to investigate if P2X7 receptor is related to the expression of klotho in the onset of diabetic nephropathy in rats. Seven-week-old male Wistar rats weighing 210 g were all uninephrectomized; two-third of the animals were induced to diabetes with 60 mg/kg streptozotocin i.v., and one-third received its vehicle (control rats). At 4th day of the fifth week of the protocol, half of the diabetic rats received a small interfering RNA targeting for P2X7 mRNA, and the other half received its vehicle. Euthanasia was made at the eighth week. Diabetic animals reproduced all classic symptoms of the disease; besides, they showed reduced renal function and low NO bioavailability; also, SOD1, SOD2, and catalase were increased, probably due to the oxidative stress which was elevated in this situation. Metabolic data of diabetic rats did not change by silencing P2X7 receptor. For the other hand, silencing P2X7 was able to contribute to balance oxidative and nitrosative profile, ultimately improving the renal function and increasing plasma and membrane forms of klotho. These findings suggest that the management of P2X7 receptor can benefit the kidneys with diabetic nephropathy. Further studies are needed to show the therapeutic potential of this receptor inhibition to provide a better quality of life for the diabetic patient.
Collapse
Affiliation(s)
- A M Rodrigues
- Translational Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - R S Serralha
- Translational Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - D Y Lima
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Nephrology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - G R Punaro
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
- Nephrology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - I Visona
- Department of Pathology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - M J S Fernandes
- Department of Neurology and Neurosurgery, Neuroscience, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - E M S Higa
- Translational Medicine, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
- Nephrology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
- Emergency Division, Department of Medicine, Universidade Federal Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
34
|
Cho KS, Lee JH, Cho J, Cha GH, Song GJ. Autophagy Modulators and Neuroinflammation. Curr Med Chem 2020; 27:955-982. [PMID: 30381067 DOI: 10.2174/0929867325666181031144605] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/20/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neuroinflammation plays a critical role in the development and progression of various neurological disorders. Therefore, various studies have focused on the development of neuroinflammation inhibitors as potential therapeutic tools. Recently, the involvement of autophagy in the regulation of neuroinflammation has drawn substantial scientific interest, and a growing number of studies support the role of impaired autophagy in the pathogenesis of common neurodegenerative disorders. OBJECTIVE The purpose of this article is to review recent research on the role of autophagy in controlling neuroinflammation. We focus on studies employing both mammalian cells and animal models to evaluate the ability of different autophagic modulators to regulate neuroinflammation. METHODS We have mostly reviewed recent studies reporting anti-neuroinflammatory properties of autophagy. We also briefly discussed a few studies showing that autophagy modulators activate neuroinflammation in certain conditions. RESULTS Recent studies report neuroprotective as well as anti-neuroinflammatory effects of autophagic modulators. We discuss the possible underlying mechanisms of action of these drugs and their potential limitations as therapeutic agents against neurological disorders. CONCLUSION Autophagy activators are promising compounds for the treatment of neurological disorders involving neuroinflammation.
Collapse
Affiliation(s)
- Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul, Korea
| | - Jang Ho Lee
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Korea
| | - Jeiwon Cho
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Korea.,Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| | - Guang-Ho Cha
- Department of Medical Science, College of Medicine, Chungnam National University, 35015 Daejeon, Korea
| | - Gyun Jee Song
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Korea.,Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| |
Collapse
|
35
|
Xie D, Ge X, Ma Y, Tang J, Wang Y, Zhu Y, Gao C, Pan S. Clemastine improves hypomyelination in rats with hypoxic-ischemic brain injury by reducing microglia-derived IL-1β via P38 signaling pathway. J Neuroinflammation 2020; 17:57. [PMID: 32061255 PMCID: PMC7023767 DOI: 10.1186/s12974-019-1662-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Microglia activation is associated with the development of hypoxic–ischemic brain injury (HIBI). Neuroinflammation suppression might be a suitable therapeutic target in hypoxic oligodendrocyte injury. This study aims to determine whether clemastine can improve hypomyelination by suppressing the activated microglia and promoting the maturation of oligodendrocyte progenitor cells (OPCs) in HIBI. Methods A bilateral common carotid artery occlusion (BCCAO) rat model that received continuous intraperitoneal injection (1 mg/kg) for 14 days was employed to elaborate the neuroprotection effects of clemastine. Interleukin-1β (IL-1β), nod-like receptor protein 3 (NLRP3), histamine H1 receptor, and OPC differentiation levels in the corpus callosum were measured. Primary cultured OPCs and co-culture of microglia and OPCs were used to explore the link between microglia activation and hypomyelination. Data were evaluated by one-way ANOVA with Fisher’s protected least significant difference test. Results Clemastine treatment could reverse hypomyelination and restrain the upregulation of IL-1β and NLRP3 in the corpus callosum of BCCAO rats. Primary cultured OPCs treated with IL-1β showed failed maturation. However, clemastine could also reverse the OPC maturation arrest by activating the extracellular signal-regulated kinase (ERK) signaling pathway. Co-culture of microglia and OPCs with oxygen glucose deprivation treatment exhibited IL-1β and NLRP3 upregulation. Clemastine could downregulate NLRP3 and IL-1β and reverse hypomyelination by inhibiting the p38 signaling pathway. Conclusions Clemastine could restrain microglia activation, improve axonal hypomyelination in BCCAO rats, and thus might be a viable strategy to inhibit hypomyelination in the corpus callosum of patients with HIBI.
Collapse
Affiliation(s)
- Di Xie
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Xiaoli Ge
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Yanli Ma
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Jialong Tang
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Yang Wang
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Yajie Zhu
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| |
Collapse
|
36
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
37
|
Filipi T, Hermanova Z, Tureckova J, Vanatko O, Anderova M. Glial Cells-The Strategic Targets in Amyotrophic Lateral Sclerosis Treatment. J Clin Med 2020; 9:E261. [PMID: 31963681 PMCID: PMC7020059 DOI: 10.3390/jcm9010261] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease, which is characterized by the degeneration of motor neurons in the motor cortex and the spinal cord and subsequently by muscle atrophy. To date, numerous gene mutations have been linked to both sporadic and familial ALS, but the effort of many experimental groups to develop a suitable therapy has not, as of yet, proven successful. The original focus was on the degenerating motor neurons, when researchers tried to understand the pathological mechanisms that cause their slow death. However, it was soon discovered that ALS is a complicated and diverse pathology, where not only neurons, but also other cell types, play a crucial role via the so-called non-cell autonomous effect, which strongly deteriorates neuronal conditions. Subsequently, variable glia-based in vitro and in vivo models of ALS were established and used for brand-new experimental and clinical approaches. Such a shift towards glia soon bore its fruit in the form of several clinical studies, which more or less successfully tried to ward the unfavourable prognosis of ALS progression off. In this review, we aimed to summarize current knowledge regarding the involvement of each glial cell type in the progression of ALS, currently available treatments, and to provide an overview of diverse clinical trials covering pharmacological approaches, gene, and cell therapies.
Collapse
Affiliation(s)
- Tereza Filipi
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Ondrej Vanatko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| |
Collapse
|
38
|
Volonté C, Apolloni S, Sabatelli M. Histamine beyond its effects on allergy: Potential therapeutic benefits for the treatment of Amyotrophic Lateral Sclerosis (ALS). Pharmacol Ther 2019; 202:120-131. [PMID: 31233766 DOI: 10.1016/j.pharmthera.2019.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
Abstract
ALS currently remains a challenge despite many efforts in performing successful clinical trials and formulating therapeutic solutions. By learning from current failures and striving for success, scientists and clinicians are checking every possibility to search for missing hints and efficacious treatments. Because the disease is very complex and heterogeneous and, moreover, targeting not only motor neurons but also several different cell types including muscle, glial, and immune cells, the right answer to ALS is conceivably a multidrug strategy or the use of broad-spectrum molecules. The aim of the present work is to gather evidence about novel perspectives on ALS pathogenesis and to present recent and innovative paradigms for therapy. In particular, we describe how an old molecule possessing immunomodulatory and neuroprotective functions beyond its recognized effects on allergy, histamine, might have a renewed and far-reaching momentum in ALS.
Collapse
Affiliation(s)
- Cinzia Volonté
- CNR-Institute of Cell Biology and Neurobiology/UCSC, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Savina Apolloni
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via Del Fosso di Fiorano 65, 00143 Rome, Italy
| | - Mario Sabatelli
- Institute of Neurology-Catholic University of Sacro Cuore, Clinic Center NEMO- Fondazione Pol. A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| |
Collapse
|
39
|
Functional microglia neurotransmitters in amyotrophic lateral sclerosis. Semin Cell Dev Biol 2019; 94:121-128. [PMID: 31009755 DOI: 10.1016/j.semcdb.2019.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
Abstract
Today neuroscience is dominated by the perspective that microglia are essential elements in any integrated view of the nervous system. A number of different neuroinflammatory conditions affect the CNS where microglia involvement, and particularly microgliosis, is not only a prominent feature, but also a pathogenic key mechanism of disease. On the other side, microglia can also constitute an important trigger of neuronal protection during neurodegenerative disorders. For instance in ALS and other motor neuron diseases, available evidence suggests the coexistence of quite different roles for microglia, characterized by neuroprotective functions at early stages, and neurotoxic actions during disease progression. The scope of this review is a brief discussion about microglia being activated and functioning during ALS, and particularly about neurotransmitters participating to the pathological signature of ALS microglia. We will discuss that ALS microglia can express a variety of classical neurotransmitter receptors comprising those for extracellular ATP, glutamate and histamine. We will review data indicating that the modulation of these transmitter receptors may induce beneficial effects in ALS models, so that the protective properties of microglia can be emphasized at the expenses of their toxicity.
Collapse
|
40
|
Su WJ, Zhang T, Jiang CL, Wang W. Clemastine Alleviates Depressive-Like Behavior Through Reversing the Imbalance of Microglia-Related Pro-inflammatory State in Mouse Hippocampus. Front Cell Neurosci 2018; 12:412. [PMID: 30483062 PMCID: PMC6243034 DOI: 10.3389/fncel.2018.00412] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022] Open
Abstract
Backgrounds: Abundant reports indicate that neuroinflammatory signaling contributes to behavioral complications associated with depression and may be related to treatment response. The glial cells, especially microglia and astrocytes in brain regions of hippocampus and medial prefrontal cortex (mPFC), are major components of CNS innate immunity. Moreover, purinergic receptor P2X, ligand-gated ion channel 7 (P2X7R) was recently reckoned as a pivotal regulator in central immune system. Besides, it was pointed out that clemastine, a first-generation histamine receptor H1 (HRH1) antagonist with considerable safety profile and pharmacological effect, may suppress immune activation through modulating P2X7R. Herein, we investigated the potential anti-neuroinflammatory effects of clemastine on chronic unpredictable mild stress (CUMS)-induced depressive-like behavior in a mouse model. Methods: Male BALB/c mice were subjected to CUMS for 4 weeks, some of them were injected with clemastine fumarate solution. After the stress procedure, behavioral tests including Sucrose Preference Tests (SPTs), Tail Suspension Tests (TSTs) and locomotor activities were performed to evaluate depressive-like phenotype. Subsequently, expression of cytokines and microglia-related inflammatory biomarkers were assessed. Results: In the present research, we found that clemastine significantly reversed both the declination of SPT percentage and the extension of TST immobility durations in depression mouse model without affecting locomotor activity. Also, we observed that clemastine regulated the imbalance of pro-inflammatory cytokines including interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in the hippocampus and serum of depressive-like mice. Additionally, clemastine significantly suppressed microglial M1-like activation specifically in the hippocampus, and also improved hippocampal astrocytic loss. Furthermore, clemastine downregulated hippocampal P2X7R without interfering with the expression of HRH1. Conclusion: As a safe and efficient anti-allergic agent, clemastine could impressively alleviate stress-related depressive-like phenotype in mice. Further evidence supported that it was because of the potential function of clemastine in modulating the expression of P2X7 receptor possibly independent of HRH1, therefore suppressing the microglial M1-like activation and pro-inflammatory cytokines release in brain regions of hippocampus rather than mPFC.
Collapse
Affiliation(s)
- Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, China
| | - Ting Zhang
- Department of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, China.,Department of Navy Aviation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, China
| | - Wei Wang
- Department of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, China
| |
Collapse
|
41
|
Cocozza G, di Castro MA, Carbonari L, Grimaldi A, Antonangeli F, Garofalo S, Porzia A, Madonna M, Mainiero F, Santoni A, Grassi F, Wulff H, D'Alessandro G, Limatola C. Ca 2+-activated K + channels modulate microglia affecting motor neuron survival in hSOD1 G93A mice. Brain Behav Immun 2018; 73:584-595. [PMID: 29981425 PMCID: PMC6129409 DOI: 10.1016/j.bbi.2018.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
Recent studies described a critical role for microglia in amyotrophic lateral sclerosis (ALS), where these CNS-resident immune cells participate in the establishment of an inflammatory microenvironment that contributes to motor neuron degeneration. Understanding the mechanisms leading to microglia activation in ALS could help to identify specific molecular pathways which could be targeted to reduce or delay motor neuron degeneration and muscle paralysis in patients. The intermediate-conductance calcium-activated potassium channel KCa3.1 has been reported to modulate the "pro-inflammatory" phenotype of microglia in different pathological conditions. We here investigated the effects of blocking KCa3.1 activity in the hSOD1G93AALS mouse model, which recapitulates many features of the human disease. We report that treatment of hSOD1G93A mice with a selective KCa3.1 inhibitor, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), attenuates the "pro-inflammatory" phenotype of microglia in the spinal cord, reduces motor neuron death, delays onset of muscle weakness, and increases survival. Specifically, inhibition of KCa3.1 channels slowed muscle denervation, decreased the expression of the fetal acetylcholine receptor γ subunit and reduced neuromuscular junction damage. Taken together, these results demonstrate a key role for KCa3.1 in driving a pro-inflammatory microglia phenotype in ALS.
Collapse
Affiliation(s)
- Germana Cocozza
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; Center for Life Nanoscience - Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | | | - Laura Carbonari
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Alfonso Grimaldi
- Center for Life Nanoscience - Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Fabrizio Antonangeli
- Department of Molecular Medicine, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Rome, Italy
| | | | | | - Fabrizio Mainiero
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Francesca Grassi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | | | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy.
| |
Collapse
|
42
|
Gupta N, Shyamasundar S, Patnala R, Karthikeyan A, Arumugam TV, Ling EA, Dheen ST. Recent progress in therapeutic strategies for microglia-mediated neuroinflammation in neuropathologies. Expert Opin Ther Targets 2018; 22:765-781. [DOI: 10.1080/14728222.2018.1515917] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Neelima Gupta
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sukanya Shyamasundar
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Radhika Patnala
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Aparna Karthikeyan
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thiruma V. Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eng-Ang Ling
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - S. Thameem Dheen
- Department of Anatomy Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
43
|
Fabbrizio P, Amadio S, Apolloni S, Volonté C. P2X7 Receptor Activation Modulates Autophagy in SOD1-G93A Mouse Microglia. Front Cell Neurosci 2017; 11:249. [PMID: 28871219 PMCID: PMC5566572 DOI: 10.3389/fncel.2017.00249] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022] Open
Abstract
Autophagy and inflammation play determinant roles in the pathogenesis of Amyotrophic Lateral Sclerosis (ALS), an adult-onset neurodegenerative disease characterized by deterioration and final loss of upper and lower motor neurons (MN) priming microglia to sustain neuroinflammation and a vicious cycle of neurodegeneration. Given that extracellular ATP through P2X7 receptor constitutes a neuron-to-microglia alarm signal implicated in ALS, and that P2X7 affects autophagy in immune cells, we have investigated if autophagy can be directly triggered by P2X7 activation in primary microglia from superoxide dismutase 1 (SOD1)-G93A mice. We report that P2X7 enhances the expression of the autophagic marker microtubule-associated protein 1 light chain 3 (LC3)-II, via mTOR pathway and concomitantly with modulation of anti-inflammatory M2 microglia markers. We also demonstrate that the autophagic target SQSTM1/p62 is decreased in SOD1-G93A microglia after a short stimulation of P2X7, but increased after a sustained challenge. These effects are prevented by the P2X7 antagonist A-804598, and the autophagy/phosphoinositide-3-kinase inhibitor wortmannin (WM). Finally, a chronic in vivo treatment with A-804598 in SOD1-G93A mice decreases the expression of SQSTM1/p62 in lumbar spinal cord at end stage of disease. These data identify the modulation of the autophagic flux as a novel mechanism by which P2X7 activates ALS-microglia, to be considered for further investigations in ALS.
Collapse
Affiliation(s)
- Paola Fabbrizio
- IRCCS Santa Lucia Foundation, Experimental NeuroscienceRome, Italy.,Department of Systems Medicine, Tor Vergata UniversityRome, Italy
| | - Susanna Amadio
- IRCCS Santa Lucia Foundation, Experimental NeuroscienceRome, Italy
| | - Savina Apolloni
- IRCCS Santa Lucia Foundation, Experimental NeuroscienceRome, Italy
| | - Cinzia Volonté
- IRCCS Santa Lucia Foundation, Experimental NeuroscienceRome, Italy.,CNR, Institute of Cell Biology and NeurobiologyRome, Italy
| |
Collapse
|
44
|
Geloso MC, Corvino V, Marchese E, Serrano A, Michetti F, D'Ambrosi N. The Dual Role of Microglia in ALS: Mechanisms and Therapeutic Approaches. Front Aging Neurosci 2017; 9:242. [PMID: 28790913 PMCID: PMC5524666 DOI: 10.3389/fnagi.2017.00242] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/11/2017] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by a non-cell autonomous motor neuron loss. While it is generally believed that the disease onset takes place inside motor neurons, different cell types mediating neuroinflammatory processes are considered deeply involved in the progression of the disease. On these grounds, many treatments have been tested on ALS animals with the aim of inhibiting or reducing the pro-inflammatory action of microglia and astrocytes and counteract the progression of the disease. Unfortunately, these anti-inflammatory therapies have been only modestly successful. The non-univocal role played by microglia during stress and injuries might explain this failure. Indeed, it is now well recognized that, during ALS, microglia displays different phenotypes, from surveillant in early stages, to activated states, M1 and M2, characterized by the expression of respectively harmful and protective genes in later phases of the disease. Consistently, the inhibition of microglial function seems to be a valid strategy only if the different stages of microglia polarization are taken into account, interfering with the reactivity of microglia specifically targeting only the harmful pathways and/or potentiating the trophic ones. In this review article, we will analyze the features and timing of microglia activation in the light of M1/M2 phenotypes in the main mice models of ALS. Moreover, we will also revise the results obtained by different anti-inflammatory therapies aimed to unbalance the M1/M2 ratio, shifting it towards a protective outcome.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro CuoreRome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro CuoreRome, Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro CuoreRome, Italy
| | - Alessia Serrano
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro CuoreRome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro CuoreRome, Italy.,IRCCS San Raffaele Scientific Institute, Università Vita-Salute San RaffaeleMilan, Italy
| | - Nadia D'Ambrosi
- Department of Biology, University of Rome Tor VergataRome, Italy
| |
Collapse
|
45
|
Kim J, Song JH. Inhibitory effects of antihistamines, diphenhydramine and chlorpheniramine, on proton currents in BV2 microglial cells. Eur J Pharmacol 2017; 798:122-128. [PMID: 28131782 DOI: 10.1016/j.ejphar.2017.01.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/02/2017] [Accepted: 01/25/2017] [Indexed: 02/08/2023]
Abstract
Microglial NADPH oxidase is a major source of toxic reactive oxygen species produced during chronic neuroinflammation. Voltage-gated proton channel (HV1) functions to maintain the intense activity of NADPH oxidase, and channel inhibition alleviates the pathology of neurodegenerative diseases such as ischemic stroke and multiple sclerosis associated with oxidative neuroinflammation. Antagonists of histamine H1 receptors have beneficial effects against microglia-mediated oxidative stress and neurotoxicity. We examined the effects of the H1 antihistamines, diphenhydramine and chlorpheniramine, on proton currents in BV2 microglial cells recorded using the whole-cell patch clamp technique. Diphenhydramine and chlorpheniramine reduced the proton currents with almost the same potency, yielding IC50 values of 42 and 43μM, respectively. Histamine did not affect proton currents, excluding the involvement of histamine receptors in their action. Neither drug shifted the voltage-dependence of activation or the reversal potential of the proton currents, even though diphenhydramine slowed the activation and deactivation kinetics. The inhibitory effects of the two antihistamines on proton currents could be utilized to develop therapeutic agents for neurodegenerative diseases and other diseases associated with HV1 proton channel abnormalities.
Collapse
Affiliation(s)
- Jiwon Kim
- Department of Pharmacology, College of Medicine, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Jin-Ho Song
- Department of Pharmacology, College of Medicine, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
46
|
Monczor F, Fernandez N. Current Knowledge and Perspectives on Histamine H1 and H2 Receptor Pharmacology: Functional Selectivity, Receptor Crosstalk, and Repositioning of Classic Histaminergic Ligands. Mol Pharmacol 2016; 90:640-648. [PMID: 27625037 DOI: 10.1124/mol.116.105981] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022] Open
Abstract
H1 and H2 histamine receptor antagonists, although developed many decades ago, are still effective for the treatment of allergic and gastric acid-related conditions. This article focuses on novel aspects of the pharmacology and molecular mechanisms of histamine receptors that should be contemplated for optimizing current therapies, repositioning histaminergic ligands for new therapeutic uses, or even including agonists of the histaminergic system in the treatment of different pathologies such as leukemia or neurodegenerative disorders. In recent years, new signaling phenomena related to H1 and H2 receptors have been described that make them suitable for novel therapeutic approaches. Crosstalk between histamine receptors and other membrane or nuclear receptors can be envisaged as a way to modulate other signaling pathways and to potentiate the efficacy of drugs acting on different receptors. Likewise, biased signaling at histamine receptors seems to be a pharmacological feature that can be exploited to investigate nontraditional therapeutic uses for H1 and H2 biased agonists in malignancies such as acute myeloid leukemia and to avoid undesired side effects when used in standard treatments. It is hoped that the molecular mechanisms discussed in this review contribute to a better understanding of the different aspects involved in histamine receptor pharmacology, which in turn will contribute to increased drug efficacy, avoidance of adverse effects, or repositioning of histaminergic ligands.
Collapse
Affiliation(s)
- Federico Monczor
- Instituto de Investigaciones Farmacológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Fernandez
- Instituto de Investigaciones Farmacológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
47
|
Apolloni S, Fabbrizio P, Amadio S, Volonté C. Actions of the antihistaminergic clemastine on presymptomatic SOD1-G93A mice ameliorate ALS disease progression. J Neuroinflammation 2016; 13:191. [PMID: 27549088 PMCID: PMC4994328 DOI: 10.1186/s12974-016-0658-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a disease with a strong neuroinflammatory component sustained by activated microglia contributing to motoneuron death. However, how to successfully balance neuroprotective versus neurotoxic actions by the use of antinflammatory agents is still under scrutiny. We have recently shown that the antihistamine clemastine, an FDA-approved drug, can influence the M1/M2 switch occurring in SOD1-G93A ALS microglia. METHODS Here, we have chronically treated female SOD1-G93A mice with clemastine, evaluated disease progression and performed mice lumbar spinal cord analysis at symptomatic and end stage of the disease. Moreover, we have studied the mechanism of action of clemastine in primary adult spinal SOD1-G93A microglia cultures and in NSC-G93A motor neuron-like cells. RESULTS We found that a short treatment with clemastine (50 mg/kg) from asymptomatic (postnatal day 40) to symptomatic phase (postnatal day 120) significantly delayed disease onset and extended the survival of SOD1-G93A mice by about 10 %. Under these conditions, clemastine induced protection of motor neurons, modulation of inflammatory parameters, reduction of SOD1 protein levels and SQSTM1/p62 autophagic marker, when analysed immediately at the end of the treatment (postnatal day 120). A long treatment with clemastine (from asymptomatic until the end stage) instead failed to ameliorate ALS disease progression. At the end stage of the disease, we found that clemastine short treatment decreased microgliosis and SOD1 protein and increased LC3-II autophagic marker, while the long treatment produced opposite effects. Finally, in spinal microglia cultures from symptomatic SOD1-G93A mice clemastine activated inflammatory parameters, stimulated autophagic flux via the mTOR signalling pathway and decreased SOD1 levels. Modulation of autophagy was also demonstrated in NSC34 SOD1-G93A motor neuron-like cells. CONCLUSIONS By gaining insights into the ameliorating actions of an antihistaminergic compound in ALS disease, our findings might represent an exploitable therapeutic approach for familial forms of ALS.
Collapse
Affiliation(s)
- Savina Apolloni
- Santa Lucia Foundation, IRCCS, Rome, Italy.,Institute of Cell Biology and Neurobiology, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy
| | | | - Susanna Amadio
- Santa Lucia Foundation, IRCCS, Rome, Italy.,Institute of Cell Biology and Neurobiology, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy
| | - Cinzia Volonté
- Santa Lucia Foundation, IRCCS, Rome, Italy. .,Institute of Cell Biology and Neurobiology, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy.
| |
Collapse
|
48
|
Lloyd AF, Miron VE. Cellular and Molecular Mechanisms Underpinning Macrophage Activation during Remyelination. Front Cell Dev Biol 2016; 4:60. [PMID: 27446913 PMCID: PMC4914869 DOI: 10.3389/fcell.2016.00060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022] Open
Abstract
Remyelination is an example of central nervous system (CNS) regeneration, whereby myelin is restored around demyelinated axons, re-establishing saltatory conduction and trophic/metabolic support. In progressive multiple sclerosis, remyelination is limited or fails altogether which is considered to contribute to axonal damage/loss and consequent disability. Macrophages have critical roles in both CNS damage and regeneration, such as remyelination. This diverse range in functions reflects the ability of macrophages to acquire tissue microenvironment-specific activation states. This activation is dynamically regulated during efficient regeneration, with a switch from pro-inflammatory to inflammation-resolution/pro-regenerative phenotypes. Although, some molecules and pathways have been implicated in the dynamic activation of macrophages, such as NFκB, the cellular and molecular mechanisms underpinning plasticity of macrophage activation are unclear. Identifying mechanisms regulating macrophage activation to pro-regenerative phenotypes may lead to novel therapeutic strategies to promote remyelination in multiple sclerosis.
Collapse
Affiliation(s)
- Amy F Lloyd
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh Edinburgh, UK
| | - Veronique E Miron
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh Edinburgh, UK
| |
Collapse
|
49
|
Rocha SM, Saraiva T, Cristóvão AC, Ferreira R, Santos T, Esteves M, Saraiva C, Je G, Cortes L, Valero J, Alves G, Klibanov A, Kim YS, Bernardino L. Histamine induces microglia activation and dopaminergic neuronal toxicity via H1 receptor activation. J Neuroinflammation 2016; 13:137. [PMID: 27260166 PMCID: PMC4893260 DOI: 10.1186/s12974-016-0600-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
Background Histamine is an amine widely known as a peripheral inflammatory mediator and as a neurotransmitter in the central nervous system. Recently, it has been suggested that histamine acts as an innate modulator of microglial activity. Herein, we aimed to disclose the role of histamine in microglial phagocytic activity and reactive oxygen species (ROS) production and to explore the consequences of histamine-induced neuroinflammation in dopaminergic (DA) neuronal survival. Methods The effect of histamine on phagocytosis was assessed both in vitro by using a murine N9 microglial cell line and primary microglial cell cultures and in vivo. Cells were exposed to IgG-opsonized latex beads or phosphatidylserine (PS) liposomes to evaluate Fcγ or PS receptor-mediated microglial phagocytosis, respectively. ROS production and protein levels of NADPH oxidases and Rac1 were assessed as a measure of oxidative stress. DA neuronal survival was evaluated in vivo by counting the number of tyrosine hydroxylase-positive neurons in the substantia nigra (SN) of mice. Results We found that histamine triggers microglial phagocytosis via histamine receptor 1 (H1R) activation and ROS production via H1R and H4R activation. By using apocynin, a broad NADPH oxidase (Nox) inhibitor, and Nox1 knockout mice, we found that the Nox1 signaling pathway is involved in both phagocytosis and ROS production induced by histamine in vitro. Interestingly, both apocynin and annexin V (used as inhibitor of PS-induced phagocytosis) fully abolished the DA neurotoxicity induced by the injection of histamine in the SN of adult mice in vivo. Blockade of H1R protected against histamine-induced Nox1 expression and death of DA neurons in vivo. Conclusions Overall, our results highlight the relevance of histamine in the modulation of microglial activity that ultimately may interfere with neuronal survival in the context of Parkinson’s disease (PD) and, eventually, other neurodegenerative diseases which are accompanied by microglia-induced neuroinflammation. Importantly, our results also open promising new perspectives for the therapeutic use of H1R antagonists to treat or ameliorate neurodegenerative processes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0600-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandra M Rocha
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Tatiana Saraiva
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ana C Cristóvão
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Raquel Ferreira
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Tiago Santos
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Marta Esteves
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cláudia Saraiva
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Goun Je
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Luísa Cortes
- Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Jorge Valero
- Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Gilberto Alves
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Alexander Klibanov
- Division of Cardiovascular Medicine and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Yoon-Seong Kim
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal. .,Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
| |
Collapse
|
50
|
Bhattacharya A, Biber K. The microglial ATP-gated ion channel P2X7 as a CNS drug target. Glia 2016; 64:1772-87. [PMID: 27219534 DOI: 10.1002/glia.23001] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/17/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022]
Abstract
Based on promising preclinical evidence, microglial P2X7 has increasingly being recognized as a target for therapeutic intervention in neurological and psychiatric diseases. However, despite this knowledge no P2X7-related drug has yet entered clinical trials with respect to CNS diseases. We here discuss the current literature on P2X7 being a drug target and identify unsolved issues and still open questions that have hampered the development of P2X7 dependent therapeutic approaches for CNS diseases. It is concluded here that the lack of brain penetrating P2X7 antagonists is a major obstacle in the field and that central P2X7 is a yet untested clinical drug target. In the CNS, microglial P2X7 activation causes neuroinflammation, which in turn plays a role in various CNS disorders. This has resulted in a surge of brain penetrant P2X7 antagonists. P2X7 is a viable, clinically untested CNS drug target. GLIA 2016;64:1772-1787.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- LLC. Neuroscience Drug Discovery, Janssen Research & Development, 3210 Merryfield Row, San Diego, California
| | - Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Hauptstrasse 5, Freiburg, Germany.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, AV Groningen, The Netherlands
| |
Collapse
|