1
|
Chen CM, Huang CY, Lai CH, Chen YC, Hwang YT, Lin CY. Neuroprotection effects of kynurenic acid-loaded micelles for the Parkinson's disease models. J Liposome Res 2024; 34:593-604. [PMID: 38779944 DOI: 10.1080/08982104.2024.2346986] [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: 01/31/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
Anti-glutamatergic agents may have neuroprotective effects against excitotoxicity that is known to be involved in the pathogenesis of Parkinson's disease (PD). One of these agents is kynurenic acid (KYNA), a tryptophan metabolite, which is an endogenous N-methyl-D-aspartic acid (NMDA) receptor antagonist. However, its pharmacological properties of poor water solubility and limited blood-brain barrier (BBB) permeability rules out its systemic administration in disorders affecting the central nervous system. Our aim in the present study was to investigate the neuroprotective effects of KYNA-loaded micelles (KYNA-MICs) against PD in vitro and in vivo. Lipid-based micelles (MICs) in conjunction with KYNA drug delivery have the potential to enhance the penetration of therapeutic drugs into a diseased brain without BBB obstacles. KYNA-MICs were characterized by particle size (105.8 ± 12.1 nm), loading efficiency (78.3 ± 4.23%), and in vitro drug release (approximately 30% at 24 h). The in vitro experiments showed that KYNA-MICs effectively reduced 2-fold protein aggregation. The in vivo studies revealed that KYNA was successfully delivered by 5-fold increase in neurotoxin-induced PD brains. The results showed significant enhancement of KYNA delivery into brain. We also found that the KYNA-MICs exhibited several therapeutic effects. The KYNA-MICs reduced protein aggregation of an in vitro PD model, ameliorated motor functions, and prevented loss of the striatal neurons in a PD animal model. The beneficial effects of KYNA-MICs are probably explained by the anti-excitotoxic activity of the treatment's complex. As the KYNA-MICs did not induce any appreciable side-effects at the protective dose applied to a chronic PD mouse model, our results demonstrate that KYNA provides neuroprotection and attenuates PD pathology.
Collapse
Affiliation(s)
- Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Yun Huang
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Hui Lai
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chieh Chen
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ting Hwang
- Department of Statistics, National Taipei University, Taipei, Taiwan
| | - Chung-Yin Lin
- Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Radiation Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Statistics, National Taipei University, Taipei, Taiwan
| |
Collapse
|
2
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 DOI: 10.3390/ijms252312637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
3
|
Takeda A, Tamano H. Insight into brain metallothioneins from bidirectional Zn2+ signaling in synaptic dynamics. Metallomics 2024; 16:mfae039. [PMID: 39223100 DOI: 10.1093/mtomcs/mfae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/01/2024] [Indexed: 09/04/2024]
Abstract
The basal levels as the labile Zn2+ pools in the extracellular and intracellular compartments are in the range of ∼10 nM and ∼100 pM, respectively. The influx of extracellular Zn2+ is used for memory via cognitive activity and is regulated for synaptic plasticity, a cellular mechanism of memory. When Zn2+ influx into neurons excessively occurs, however, it becomes a critical trigger for cognitive decline and neurodegeneration, resulting in acute and chronic pathogenesis. Aging, a biological process, generally accelerates vulnerability to neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). The basal level of extracellular Zn2+ is age relatedly increased in the rat hippocampus, and the influx of extracellular Zn2+ contributes to accelerating vulnerability to the AD and PD pathogenesis in experimental animals with aging. Metallothioneins (MTs) are Zn2+-binding proteins for cellular Zn2+ homeostasis and involved in not only supplying functional Zn2+ required for cognitive activity, but also capturing excess (toxic) Zn2+ involved in cognitive decline and neurodegeneration. Therefore, it is estimated that regulation of MT synthesis is involved in both neuronal activity and neuroprotection. The present report provides recent knowledge regarding the protective/preventive potential of MT synthesis against not only normal aging but also the AD and PD pathogenesis in experimental animals, focused on MT function in bidirectional Zn2+ signaling in synaptic dynamics.
Collapse
Affiliation(s)
- Atsushi Takeda
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Shizuoka Tohto Medical College, 1949 Minamiema, Izunokuni, Shizuoka 410-2221, Japan
| |
Collapse
|
4
|
Wang Y, Xu H, Chen S, Chen J, Zheng Q, Ma Y, Zhao X, Shi Y, Xiao L. Oxytocin Protects Nigrostriatal Dopamine Signal via Activating GABAergic Circuit in the MPTP-Induced Parkinson's Disease Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310244. [PMID: 39099429 PMCID: PMC11423065 DOI: 10.1002/advs.202310244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/23/2024] [Indexed: 08/06/2024]
Abstract
The most pronounced neuropathological feature of Parkinson's disease (PD) is the loss of dopamine (DA) neurons in the substantia nigra compacta (SNc), which depletes striatal DA. Hypothalamic oxytocin is found to be reduced in PD patients and closely interacts with the DA system, but the role of oxytocin in PD remains unclear. Here, the disturbances of endogenous oxytocin level and the substantia nigra (SN) oxytocin receptor expression in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model is observed, correlated with the striatal tyrosine hydroxylase (TH) expression reduction. Killing/silencing hypothalamic oxytocin neurons aggravates the vulnerability of nigrostriatal DA signal to MPTP, whereas elevating oxytocin level by intranasal delivery or microinjecting into the SN promotes the resistance. In addition, knocking out SN oxytocin receptors induces the time-dependent reductions of SNc DA neurons, striatal TH expression, and striatal DA level by increasing neuronal excitotoxicity. These results further uncover that oxytocin dampens the excitatory synaptic inputs onto DA neurons via activating oxytocin receptor-expressed SN GABA neurons, which target GABA(B) receptors expressed in SNc DA neuron-projecting glutamatergic axons, to reduce excitotoxicity. Thus, besides the well-known prosocial effect, oxytocin acts as a key endogenous factor in protecting the nigrostriatal DA system.
Collapse
Affiliation(s)
- Yurong Wang
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hao Xu
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Saiyong Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Junhao Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qimeng Zheng
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xinru Zhao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying Shi
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
5
|
Ribarič S. The Contribution of Type 2 Diabetes to Parkinson's Disease Aetiology. Int J Mol Sci 2024; 25:4358. [PMID: 38673943 PMCID: PMC11050090 DOI: 10.3390/ijms25084358] [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: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are chronic disorders that have a significant health impact on a global scale. Epidemiological, preclinical, and clinical research underpins the assumption that insulin resistance and chronic inflammation contribute to the overlapping aetiologies of T2D and PD. This narrative review summarises the recent evidence on the contribution of T2D to the initiation and progression of PD brain pathology. It also briefly discusses the rationale and potential of alternative pharmacological interventions for PD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
Khazaal HT, El-Sayed EK, Mansour YE, Ibrahim RR, Bishr M, El Dib RA, Soliman HSM. Neuroprotective activity of Colocasia esculenta (L.) Schott leaves against monosodium glutamate-induced excitotoxicity in rats: phytochemical and molecular docking study. Nat Prod Res 2024:1-9. [PMID: 38606753 DOI: 10.1080/14786419.2024.2340061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
Colocasia esculenta (L.) Schott is a food crop with long history of use in treatment of various disorders including neurological diseases. The methanolic leaves extract (ME) and its n-butanol fraction (n-BF) demonstrated significant in vivo neuroprotective activity in monosodium glutamate induced excitotoxicity in rats. Sixteen and fifteen polyphenolic compounds were identified in n-BF and ME, respectively, using HPLC. Phytochemical investigation of n-BF followed by 1D (1H and 13C NMR) spectroscopic analyses led to isolation and identification of daucosterol (1), thermopsoside (2) and chrysoeriol 7-O-β-D-neohesperidoside (3) for the first time from genus Colocasia, in addition to orientin (4). LC/MS/MRM analysis of fraction V obtained from n-BF revealed identification of 13 polyphenolic compounds. Molecular docking of isolated compounds confirmed binding of all compounds at the target pocket with higher energy than crystallised ligand. The current study evaluated and confirmed the mechanistic aspects of neuroprotective activity of C. esculenta leaves for the first time.
Collapse
Affiliation(s)
- Heba T Khazaal
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
| | - Elsayed K El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
| | - Yara E Mansour
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
| | - Reham R Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
| | - Mokhtar Bishr
- Plant General Manager and Technical Director of the Arab Company for Pharmaceuticals and Medicinal, Plants, Cairo, Egypt
| | - Rabab A El Dib
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
| | - Hesham S M Soliman
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, Egypt
- PharmD program, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, Alexandria, Egypt
| |
Collapse
|
7
|
Izidoro Ribeiro R, Almeida Carvalho G, Almeida Chiareli R, Vieira de Assis Lima I, Quaglio Bellozi PM, Oliveira-Lima OC, Oliveira Giacomelli Á, Birbrair A, Santiago Gomez R, Pinheiro de Oliveira AC, Ulrich H, Cunha Xavier Pinto M. Glycine transporter-1 inhibition by NFPS promotes neuroprotection against striatal damage models. Neurosci Lett 2024; 826:137715. [PMID: 38460902 DOI: 10.1016/j.neulet.2024.137715] [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: 12/24/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
The striatum, an essential component of the brain's motor and reward systems, plays a pivotal role in a wide array of cognitive processes. Its dysfunction is a hallmark of neurodegenerative diseases like Parkinson's disease (PD) and Huntington's disease (HD), leading to profound motor and cognitive deficits. These conditions are often related to excitotoxicity, primarily due to overactivation of NMDA receptors (NMDAR). In the synaptic cleft, glycine transporter type 1 (GlyT1) controls the glycine levels, a NMDAR co-agonist, which modulates NMDAR function. This research explored the neuroprotective potential of NFPS, a GlyT1 inhibitor, in murine models of striatal injury. Employing models of neurotoxicity induced by 6-hydroxydopamine (PD model) and quinolinic acid (HD model), we assessed the effectiveness of NFPS pre-treatment in maintaining the integrity of striatal neurons and averting neuronal degeneration. The results indicated that NFPS pre-treatment conferred significant neuroprotection, reducing neuronal degeneration, protecting dopaminergic neurons, and preserving dendritic spines within the striatum. Additionally, this pre-treatment notably mitigated motor impairments resulting from striatal damage. The study revealed that GlyT1 inhibition led to substantial changes in the ratios of NMDAR subunits GluN2A/GluN1 and GluN2B/GluN1, 24 h after NFPS treatment. These findings underscore the neuroprotective efficacy of GlyT1 inhibition, proposing it as a viable therapeutic strategy for striatum-related damage.
Collapse
Affiliation(s)
- Raul Izidoro Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Gustavo Almeida Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Raphaela Almeida Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Isabel Vieira de Assis Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Paula Maria Quaglio Bellozi
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | | | - Alexander Birbrair
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Renato Santiago Gomez
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo-SP, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil.
| |
Collapse
|
8
|
Rademacher K, Nakamura K. Role of dopamine neuron activity in Parkinson's disease pathophysiology. Exp Neurol 2024; 373:114645. [PMID: 38092187 DOI: 10.1016/j.expneurol.2023.114645] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023]
Abstract
Neural activity is finely tuned to produce normal behaviors, and disruptions in activity likely occur early in the course of many neurodegenerative diseases. However, how neural activity is altered, and how these changes influence neurodegeneration is poorly understood. Here, we focus on evidence that the activity of dopamine neurons is altered in Parkinson's disease (PD), either as a compensatory response to degeneration or as a result of circuit dynamics or pathologic proteins, based on available human data and studies in animal models of PD. We then discuss how this abnormal activity may augment other neurotoxic phenomena in PD, including mitochondrial deficits, protein aggregation and spread, dopamine toxicity, and excitotoxicity. A more complete picture of how activity is altered and the resulting effects on dopaminergic neuron health and function may inform future therapeutic interventions to target and protect dopamine neurons from degeneration.
Collapse
Affiliation(s)
- Katerina Rademacher
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, California, 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.; Graduate Program in Neuroscience, University of California San Francisco, San Francisco, California, 94158, USA
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, California, 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.; Graduate Program in Neuroscience, University of California San Francisco, San Francisco, California, 94158, USA; Graduate Program in Biomedical Sciences, University of California San Francisco, San Francisco, California, 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, California, 94158, USA.
| |
Collapse
|
9
|
Yildirim-Balatan C, Fenyi A, Besnault P, Gomez L, Sepulveda-Diaz JE, Michel PP, Melki R, Hunot S. Parkinson's disease-derived α-synuclein assemblies combined with chronic-type inflammatory cues promote a neurotoxic microglial phenotype. J Neuroinflammation 2024; 21:54. [PMID: 38383421 PMCID: PMC10882738 DOI: 10.1186/s12974-024-03043-5] [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: 12/08/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024] Open
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by the aggregation of α-Synuclein (αSYN) building up intraneuronal inclusions termed Lewy pathology. Mounting evidence suggests that neuron-released αSYN aggregates could be central to microglial activation, which in turn mounts and orchestrates neuroinflammatory processes potentially harmful to neurons. Therefore, understanding the mechanisms that drive microglial cell activation, polarization and function in PD might have important therapeutic implications. Here, using primary microglia, we investigated the inflammatory potential of pure αSYN fibrils derived from PD patients. We further explored and characterized microglial cell responses to a chronic-type inflammatory stimulation combining PD patient-derived αSYN fibrils (FPD), Tumor necrosis factor-α (TNFα) and prostaglandin E2 (PGE2) (TPFPD). We showed that FPD hold stronger inflammatory potency than pure αSYN fibrils generated de novo. When combined with TNFα and PGE2, FPD polarizes microglia toward a particular functional phenotype departing from FPD-treated cells and featuring lower inflammatory cytokine and higher glutamate release. Whereas metabolomic studies showed that TPFPD-exposed microglia were closely related to classically activated M1 proinflammatory cells, notably with similar tricarboxylic acid cycle disruption, transcriptomic analysis revealed that TPFPD-activated microglia assume a unique molecular signature highlighting upregulation of genes involved in glutathione and iron metabolisms. In particular, TPFPD-specific upregulation of Slc7a11 (which encodes the cystine-glutamate antiporter xCT) was consistent with the increased glutamate response and cytotoxic activity of these cells toward midbrain dopaminergic neurons in vitro. Together, these data further extend the structure-pathological relationship of αSYN fibrillar polymorphs to their innate immune properties and demonstrate that PD-derived αSYN fibrils, TNFα and PGE2 act in concert to drive microglial cell activation toward a specific and highly neurotoxic chronic-type inflammatory phenotype characterized by robust glutamate release and iron retention.
Collapse
Affiliation(s)
- Cansu Yildirim-Balatan
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Alexis Fenyi
- CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, MIRCen, 92265, Fontenay-aux-Roses, France
| | - Pierre Besnault
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Lina Gomez
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Julia E Sepulveda-Diaz
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Patrick P Michel
- Sorbonne Université, Paris, France
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France
- Inserm UMRS 1127, Paris, France
- CNRS UMR 7225, Paris, France
| | - Ronald Melki
- CEA and Laboratory of Neurodegenerative Diseases, CNRS, Institut François Jacob, MIRCen, 92265, Fontenay-aux-Roses, France
| | - Stéphane Hunot
- Sorbonne Université, Paris, France.
- Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de la Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013, Paris, France.
- Inserm UMRS 1127, Paris, France.
- CNRS UMR 7225, Paris, France.
| |
Collapse
|
10
|
Zald DH. The influence of dopamine autoreceptors on temperament and addiction risk. Neurosci Biobehav Rev 2023; 155:105456. [PMID: 37926241 PMCID: PMC11330662 DOI: 10.1016/j.neubiorev.2023.105456] [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: 07/31/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
As a major regulator of dopamine (DA), DA autoreceptors (DAARs) exert substantial influence over DA-mediated behaviors. This paper reviews the physiological and behavioral impact of DAARs. Individual differences in DAAR functioning influences temperamental traits such as novelty responsivity and impulsivity, both of which are associated with vulnerability to addictive behavior in animal models and a broad array of externalizing behaviors in humans. DAARs additionally impact the response to psychostimulants and other drugs of abuse. Human PET studies of D2-like receptors in the midbrain provide evidence for parallels to the animal literature. These data lead to the proposal that weak DAAR regulation is a risk factor for addiction and externalizing problems. The review highlights the potential to build translational models of the functional role of DAARs in behavior. It also draws attention to key limitations in the current literature that would need to be addressed to further advance a weak DAAR regulation model of addiction and externalizing risk.
Collapse
Affiliation(s)
- David H Zald
- Center for Advanced Human Brain Imaging and Department of Psychiatry, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
11
|
Kumaria A, Ashkan K. Novel therapeutic strategies in glioma targeting glutamatergic neurotransmission. Brain Res 2023; 1818:148515. [PMID: 37543066 DOI: 10.1016/j.brainres.2023.148515] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/11/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023]
Abstract
High grade gliomas carry a poor prognosis despite aggressive surgical and adjuvant approaches including chemoradiotherapy. Recent studies have demonstrated a mitogenic association between neuronal electrical activity and glioma growth involving the PI3K-mTOR pathway. As the predominant excitatory neurotransmitter of the brain, glutamate signalling in particular has been shown to promote glioma invasion and growth. The concept of the neurogliomal synapse has been established whereby glutamatergic receptors on glioma cells have been shown to promote tumour propagation. Targeting glutamatergic signalling is therefore a potential treatment option in glioma. Antiepileptic medications decrease excess neuronal electrical activity and some may possess anti-glutamate effects. Although antiepileptic medications continue to be investigated for an anti-glioma effect, good quality randomised trial evidence is lacking. Other pharmacological strategies that downregulate glutamatergic signalling include riluzole, memantine and anaesthetic agents. Neuromodulatory interventions possessing potential anti-glutamate activity include deep brain stimulation and vagus nerve stimulation - this contributes to the anti-seizure efficacy of the latter and the possible neuroprotective effect of the former. A possible role of neuromodulation as a novel anti-glioma modality has previously been proposed and that hypothesis is extended to include these modalities. Similarly, the significant survival benefit in glioblastoma attributable to alternating electrical fields (Tumour Treating Fields) may be a result of disruption to neurogliomal signalling. Further studies exploring excitatory neurotransmission and glutamatergic signalling and their role in glioma origin, growth and propagation are therefore warranted.
Collapse
Affiliation(s)
- Ashwin Kumaria
- Department of Neurosurgery, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK.
| | | |
Collapse
|
12
|
Keimasi M, Salehifard K, Hoseini SJ, Esmaeili F, Esfahani NMJ, Amirsadri M, Keimasi M, Moradmand M, Mofid MR. Purified Native Protein Extracted from the Venom of Agelena orientalis Attenuates Memory Defects in the Rat Model of Glutamate-Induced Excitotoxicity. Protein J 2023; 42:586-595. [PMID: 37531037 DOI: 10.1007/s10930-023-10140-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
Spider venom contains various peptides and proteins, which can be used for pharmacological applications. Finding novel therapeutic strategies against neurodegenerative diseases with the use of purified peptides and proteins, extracted from spiders can be greatly precious. Neurodegenerative diseases are rapidly developing and expanding all over the world. Excitotoxicity is a frequent condition amongst neuro-degenerative disorders. This harmful process is usually induced through hyper-activation of N-Methyl-D-Aspartate (NMDA) receptor, and P/Q-type voltage-gated calcium channels (VGCCs). The omega-agatoxin-Aa4b is a selective and strong VGCCblocker. This study aimed to investigate the effects of this blocker on the NMDA-induced memory and learning defect in rats. For this purpose, nineteen spiders of the funnel-weaver Agelena orientalis species were collected. The extracted venom was lyophilized andpurified through gel-filtration chromatography, and capillary electrophoresis techniques. Subsequently, mass spectrometry (HPLC-ESI-MS) was used for identification of this bio-active small protein. Afterward, the effect of the omega-agatoxin-Aa4b (2 μg, intra-cornu ammonis-3 of the hippocampus) on the NMDA-induced learning and memory deficits in rats was evaluated. Learning and memory performances were evaluated by the use of passive avoidance test. For synaptic quantification and memory function the amount of calcium/calmodulin-dependent protein kinase ІІ (CaCdPKІІ) gene expression was measured using the Real-time PCR technique. To compare the experimental groups, hematoxylin and eosin (H&E) staining of hippocampus tissues was performed. Our results rendered that the omega-Agatoxin-Aa4b treatment can ameliorate and reverse the learning and memory impairment caused by NMDA-induced excitotoxicity in rat hippocampus.
Collapse
Affiliation(s)
- Mohammad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayyed Jafar Hoseini
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Noushin Mirshah Jafar Esfahani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Amirsadri
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadjavad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Moradmand
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran.
| | - Mohammad Reza Mofid
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
13
|
Serra M, Di Maio A, Bassareo V, Nuzzo T, Errico F, Servillo F, Capasso M, Parekh P, Li Q, Thiolat ML, Bezard E, Calabresi P, Sulzer D, Carta M, Morelli M, Usiello A. Perturbation of serine enantiomers homeostasis in the striatum of MPTP-lesioned monkeys and mice reflects the extent of dopaminergic midbrain degeneration. Neurobiol Dis 2023; 184:106226. [PMID: 37451474 DOI: 10.1016/j.nbd.2023.106226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Loss of dopaminergic midbrain neurons perturbs l-serine and d-serine homeostasis in the post-mortem caudate putamen (CPu) of Parkinson's disease (PD) patients. However, it is unclear whether the severity of dopaminergic nigrostriatal degeneration plays a role in deregulating serine enantiomers' metabolism. Here, through high-performance liquid chromatography (HPLC), we measured the levels of these amino acids in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys and MPTP-plus-probenecid (MPTPp)-treated mice to determine whether and how dopaminergic midbrain degeneration affects the levels of serine enantiomers in various basal ganglia subregions. In addition, in the same brain regions, we measured the levels of key neuroactive amino acids modulating glutamatergic neurotransmission, including l-glutamate, glycine, l-aspartate, d-aspartate, and their precursors l-glutamine, l-asparagine. In monkeys, MPTP treatment produced severe denervation of nigrostriatal dopaminergic fibers (⁓75%) and increased the levels of serine enantiomers in the rostral putamen (rPut), but not in the subthalamic nucleus, and the lateral and medial portion of the globus pallidus. Moreover, this neurotoxin significantly reduced the protein expression of the astrocytic serine transporter ASCT1 and the glycolytic enzyme GAPDH in the rPut of monkeys. Conversely, concentrations of d-serine and l-serine, as well as ASCT1 and GAPDH expression were unaffected in the striatum of MPTPp-treated mice, which showed only mild dopaminergic degeneration (⁓30%). These findings unveil a link between the severity of dopaminergic nigrostriatal degeneration and striatal serine enantiomers concentration, ASCT1 and GAPDH expression. We hypothesize that the up-regulation of d-serine and l-serine levels occurs as a secondary response within a homeostatic loop to support the metabolic and neurotransmission demands imposed by the degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Anna Di Maio
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Tommaso Nuzzo
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesco Errico
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy
| | - Federica Servillo
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy
| | - Mario Capasso
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Via Pansini, 5, Napoli 80131, Italy
| | - Pathik Parekh
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Qin Li
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Marie-Laure Thiolat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Erwan Bezard
- Motac Neuroscience, UKM15 6WE, Manchester, United Kingdom; Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China; Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France; Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Paolo Calabresi
- Department of Neuroscience, Cattolica Sacro Cuore University, Rome, Italy; Neurologia, Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - David Sulzer
- Departments of Psychiatry, Neurology, Pharmacology, Columbia University Irving Medical Center, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Alessandro Usiello
- Laboratory of Translational Neuroscience, CEINGE Biotecnologie Avanzate Francesco Salvatore, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università Degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
14
|
Cheng A, Zhang Y, Sun J, Huang D, Sulaiman JE, Huang X, Wu L, Ye W, Wu C, Lam H, Shi Y, Qian PY. Pterosin sesquiterpenoids from Pteris laeta Wall. ex Ettingsh. protect cells from glutamate excitotoxicity by modulating mitochondrial signals. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116308. [PMID: 36822346 DOI: 10.1016/j.jep.2023.116308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The genus Pteris (Pteridaceae) has been used as a traditional herb for a long time. In particular, Pteris laeta Wall. ex Ettingsh. has been widely used in traditional Chinese medicine to treat nervous system diseases and some pterosin sesquiterpenes from Pteris show neuroprotective activity, but their underlying molecular mechanisms remain elusive. Therefore, to investigate the neuroprotective activity and working mechanism of pterosin sesquiterpenes from P. laeta Wall. ex Ettingsh. will provide a better understanding and guidance in using P. laeta Wall. ex Ettingsh. as a traditional Chinese medicine. AIM OF THE STUDY We aim to develop effective treatments for neurodegenerative diseases from pterosin sesquiterpenes by evaluating their neuroprotective activity and investigating their working mechanisms. MATERIALS AND METHODS Primary screening on the glutamate-induced excitotoxicity cell model was assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay. Fluorescent-activated cell sorting (FACS) was used to analyze the activation level of glutamate receptors and mitochondria membrane potential after treatment. Transcriptomics and proteomics analysis was performed to identify possible targets of pterosin B. The key pathways were enriched by the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis through the Database for Annotation, Visualization, and Integrated Discovery (DAVID). The core targets were visualized by a protein-protein interaction network using STRING. The mRNA and protein expressions were evaluated using real-time quantitative polymerase chain reaction (Q-PCR) and western blot, respectively. Immunocytochemistry was performed to monitor mitochondrial and apoptotic proteins. Cellular reactive oxygen species (ROS) were measured by ROS assay, and Ca2+ was stained with Fluo-4 AM to quantify intracellular Ca2+ levels. RESULTS We found pterosin B from Pteris laeta Wall. ex Ettingsh. showed significant neuroprotective activity against glutamate excitotoxicity, enhancing cell viability from 43.8% to 105% (p-value: <0.0001). We demonstrated that pterosin B worked on the downstream signaling pathways of glutamate excitotoxicity rather than directly blocking the activation of glutamate receptors. Pterosin B restored mitochondria membrane potentials, alleviated intracellular calcium overload from 107.4% to 95.47% (p-value: 0.0006), eliminated cellular ROS by 36.55% (p-value: 0.0143), and partially secured cells from LPS-induced inflammation by increasing cell survival from 46.75% to 58.5% (p-value: 0.0114). Notably, pterosin B enhanced the expression of nuclear factor-erythroid factor 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) by 2.86-fold (p-value: 0.0006) and 4.24-fold (p-value: 0.0012), and down-regulated Kelch-like ECH-associated protein 1 (KEAP1) expression by 2.5-fold (p-value: 0.0107), indicating that it possibly promotes mitochondrial biogenesis and mitophagy to maintain mitochondria quality control and homeostasis, and ultimately inhibits apoptotic cell death. CONCLUSIONS Our work revealed that pterosin B protected cells from glutamate excitotoxicity by targeting the downstream mitochondrial signals, making it a valuable candidate for developing potential therapeutic agents in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Aifang Cheng
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Yan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China
| | - Jin Sun
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Duli Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Jordy Evan Sulaiman
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Xin Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Long Wu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Wenkang Ye
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China; SZU-HKUST Joint Ph.D. Program in Marine Environmental Science, Shenzhen University, Shenzhen, 518060, China
| | - Chuanhai Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, 999077, China
| | - Yusheng Shi
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China; Academy of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, 999077, China.
| |
Collapse
|
15
|
Pauletti C, Locuratolo N, Mannarelli D, Maffucci A, Petritis A, Menini E, Fattapposta F. Fatigue in fluctuating Parkinson's disease patients: possible impact of safinamide. J Neural Transm (Vienna) 2023:10.1007/s00702-023-02654-1. [PMID: 37210459 DOI: 10.1007/s00702-023-02654-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/13/2023] [Indexed: 05/22/2023]
Abstract
Fatigue is a common non-motor symptom in Parkinson's disease (PD). Among other pathophysiological mechanisms, neuroinflammation, a pathological PD hallmark associated with changes in glutamatergic transmission in basal ganglia, has been proposed as a crucial factor closely related to fatigue. To test the hypothesis that safinamide could represent an effective treatment of fatigue in PD patients, given its dual mechanism of action (it selectively and reversibly inhibits MAOB and modulates glutamate release), we administered the validated versions of fatigue severity scale (FSS) and Parkinson fatigue scale-16 (PFS-16) to 39 fluctuating PD patients with fatigue before and after a 24-week treatment period with safinamide as add-on therapy. An assessment of secondary variables such as depression, quality of life (QoL), and motor and non-motor symptoms (NMS) was conducted. After 24 weeks of treatment with safinamide, both FSS (p < 0.001) and PF-S16 (p = 0.02) scores were significantly lower than at baseline. Moreover, 46.2% and 41% of patients scored below the cut-off for the presence of fatigue according to FSS and PFS-16, respectively (responders). At follow-up, a significant difference emerged between responders and non-responders in mood, QoL, and NMS. Fatigue improved in fluctuating PD, and more than 40% of patients were "fatigue-free" after a 6 month treatment with safinamide. Patients without fatigue at follow-up displayed significantly better scores in QoL domains, such as mobility or activities of daily living, although disease severity remained stable, supporting the hypothesis that fatigue could considerably affect QoL. Drugs that interact with multiple neurotransmission systems, such as safinamide, could be useful in reducing this symptom.
Collapse
Affiliation(s)
- Caterina Pauletti
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy.
| | - Nicoletta Locuratolo
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
- National Centre for Disease Prevention and Health Promotion, National Institute of Health, Rome, Italy
| | - Daniela Mannarelli
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
| | - Andrea Maffucci
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
| | - Alessia Petritis
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
| | - Elisa Menini
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Francesco Fattapposta
- Department of Human Neurosciences, Universita degli Studi di Roma La Sapienza, Rome, Italy
| |
Collapse
|
16
|
Dai C, Tan C, Zhao L, Liang Y, Liu G, Liu H, Zhong Y, Liu Z, Mo L, Liu X, Chen L. Glucose Metabolism Impairment in Parkinson's Disease. Brain Res Bull 2023; 199:110672. [PMID: 37210012 DOI: 10.1016/j.brainresbull.2023.110672] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/19/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Impairments in systematic and regional glucose metabolism exist in patients with Parkinson's disease (PD) at every stage of the disease course, and such impairments are associated with the incidence, progression, and special phenotypes of PD, which affect each physiological process of glucose metabolism including glucose uptake, glycolysis, tricarboxylic acid cycle, oxidative phosphorylation, and pentose phosphate shunt pathway. These impairments may be attributed to various mechanisms, such as insulin resistance, oxidative stress, abnormal glycated modification, blood-brain-barrier dysfunction, and hyperglycemia-induced damages. These mechanisms could subsequently cause excessive methylglyoxal and reactive oxygen species production, neuroinflammation, abnormal aggregation of protein, mitochondrial dysfunction, and decreased dopamine, and finally result in energy supply insufficiency, neurotransmitter dysregulation, aggregation and phosphorylation of α-synuclein, and dopaminergic neuron loss. This review discusses the glucose metabolism impairment in PD and its pathophysiological mechanisms, and briefly summarized the currently-available therapies targeting glucose metabolism impairment in PD, including glucagon-likepeptide-1 (GLP-1) receptor agonists and dual GLP-1/gastric inhibitory peptide receptor agonists, metformin, and thiazoledinediones.
Collapse
Affiliation(s)
- Chengcheng Dai
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lili Zhao
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Yi Liang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Guohui Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Hang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Yuke Zhong
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Zhihui Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lijuan Mo
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
17
|
Tamura H, Sasaki M, Nakajima S, Nishio R, Saeki N, Katahira M, Tamano H, Takeda A. Reactive oxygen species produced by Zn 2+ influx after exposure to AMPA, but not NMDA and their capturing effect on nigral dopaminergic protection. Neurotoxicology 2023; 95:173-180. [PMID: 36775207 DOI: 10.1016/j.neuro.2023.02.003] [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: 06/22/2022] [Revised: 12/14/2022] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Glutamate excitotoxicity is involved in dopaminergic degeneration in the substantia nigra pars compacta (SNpc). Here we compared vulnerability to neurodegeneration after exposure to NMDA and AMPA. Apomorphine-induced movement disorder and dopaminergic degeneration in the SNpc, which are associated with Parkinson's syndrome, were induced after injection of AMPA into the SNpc of rats, but not after injection of NMDA. Co-injection of 1-naphthyl acetyl spermine (NASPM), a selective blocker of Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptors rescued dopaminergic degeneration and increase in intracellular Zn2+ by AMPA. Furthermore, we tested the effect of capturing reactive oxygen species (ROS) produced by Zn2+ on neuroprotection in vivo. The levels of ROS, which were determined by HYDROP, a membrane-permeable H2O2 fluorescence probe and Aminophenyl Fluorescein (APF), a fluorescence probe for hydroxyl radical and peroxynitrite, were increased after injection of AMPA, but not after co-injection of CaEDTA, an extracellular Zn2+ chelator, suggesting that increase in Zn2+ influx by AMPA elevates the levels of intracellular ROS. AMPA-mediated dopaminergic degeneration was completely rescued by co-injection of either HYDROP or APF. The present study indicates that neurotoxic signaling of the influx of extracellular Zn2+ through Zn2+-permeable GluR2-lacking AMPA receptors is converted to ROS production and that capturing the ROS completely protects dopaminergic degeneration after exposure to AMPA, but not NMDA. It is likely that regulation of the conversion from Zn2+ influx into ROS production plays a key role to preventing Parkinson's syndrome.
Collapse
Affiliation(s)
- Haruna Tamura
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Miki Sasaki
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Satoko Nakajima
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryusuke Nishio
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Nana Saeki
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Misa Katahira
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
18
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
19
|
Ishida K, Takeda K, Takehara Y, Takabayashi T, Miyara M, Sanoh S, Kawai H, Ohta S, Kotake Y. Methylmercury Decreases AMPA Receptor Subunit GluA2 Levels in Cultured Rat Cortical Neurons. Biol Pharm Bull 2023; 46:292-300. [PMID: 36724957 DOI: 10.1248/bpb.b22-00744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Methylmercury (MeHg) is a well-known environmental pollutant that has harmful effects on the central nervous systems of humans and animals. The molecular mechanisms of MeHg-induced neurotoxicity at low concentrations are not fully understood. Here, we investigated the effects of low-concentration MeHg on the cell viability, Ca2+ homeostasis, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels, which determine Ca2+ permeability of AMPA receptors, in rat primary cortical neurons. Exposure of cortical neurons to 100 and 300 nM MeHg for 7 d resulted in a decrease in GluA2 levels, an increase in basal intracellular Ca2+ concentration, increased phosphorylation levels of extracellular signal-regulated kinase (ERK)1/2 and p38, and decreased cell viability. Moreover, glutamate stimulation exacerbated the decrease in cell viability and increased intracellular Ca2+ levels in MeHg-treated neurons compared to control neurons. MeHg-induced neuronal cell death was ameliorated by 1-naphthyl acetyl spermine, a specific antagonist of Ca2+-permeable, GluA2-lacking AMPA receptors. Our findings raise the possibility that decreased neuronal GluA2 levels and the subsequent increase in intracellular Ca2+ concentration may contribute to MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Keishi Ishida
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Kazuki Takeda
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yuki Takehara
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | | | - Masatsugu Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,Wakayama Medical University
| | - Hidehiko Kawai
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shigeru Ohta
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,Wakayama Medical University
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
20
|
Milosavljević F, Brusini I, Atanasov A, Manojlović M, Vučić M, Oreščanin-Dušić Z, Brkljačić J, Miljević Č, Nikolić-Kokić A, Blagojević D, Wang C, Damberg P, Pešić V, Tyndale RF, Ingelman-Sundberg M, Jukić MM. The humanised CYP2C19 transgenic mouse exhibits cerebellar atrophy and movement impairment reminiscent of ataxia. Neuropathol Appl Neurobiol 2023; 49:e12867. [PMID: 36536486 PMCID: PMC10108232 DOI: 10.1111/nan.12867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 12/24/2022]
Abstract
AIMS CYP2C19 transgenic mouse expresses the human CYP2C19 gene in the liver and developing brain, and it exhibits altered neurodevelopment associated with impairments in emotionality and locomotion. Because the validation of new animal models is essential for the understanding of the aetiology and pathophysiology of movement disorders, the objective was to characterise motoric phenotype in CYP2C19 transgenic mice and to investigate its validity as a new animal model of ataxia. METHODS The rotarod, paw-print and beam-walking tests were utilised to characterise the motoric phenotype. The volumes of 20 brain regions in CYP2C19 transgenic and wild-type mice were quantified by 9.4T gadolinium-enhanced post-mortem structural neuroimaging. Antioxidative enzymatic activity was quantified biochemically. Dopaminergic alterations were characterised by chromatographic quantification of concentrations of dopamine and its metabolites and by subsequent immunohistochemical analyses. The beam-walking test was repeated after the treatment with dopamine receptor antagonists ecopipam and raclopride. RESULTS CYP2C19 transgenic mice exhibit abnormal, unilateral ataxia-like gait, clasping reflex and 5.6-fold more paw-slips in the beam-walking test; the motoric phenotype was more pronounced in youth. Transgenic mice exhibited a profound reduction of 12% in cerebellar volume and a moderate reduction of 4% in hippocampal volume; both regions exhibited an increased antioxidative enzyme activity. CYP2C19 mice were hyperdopaminergic; however, the motoric impairment was not ameliorated by dopamine receptor antagonists, and there was no alteration in the number of midbrain dopaminergic neurons in CYP2C19 mice. CONCLUSIONS Humanised CYP2C19 transgenic mice exhibit altered gait and functional motoric impairments; this phenotype is likely caused by an aberrant cerebellar development.
Collapse
Affiliation(s)
- Filip Milosavljević
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Irene Brusini
- Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Huddinge, Sweden.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
| | - Andrea Atanasov
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Marina Manojlović
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Marija Vučić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Zorana Oreščanin-Dušić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Brkljačić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Čedo Miljević
- Department of Psychiatry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Institute for Mental Health, Belgrade, Serbia
| | - Aleksandra Nikolić-Kokić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Duško Blagojević
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Chunliang Wang
- Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Huddinge, Sweden
| | - Peter Damberg
- Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, Solna, Sweden
| | - Vesna Pešić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Magnus Ingelman-Sundberg
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marin M Jukić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.,Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Fan Y, Huang H, Shao J, Huang W. MicroRNA-mediated regulation of reactive astrocytes in central nervous system diseases. Front Mol Neurosci 2023; 15:1061343. [PMID: 36710937 PMCID: PMC9877358 DOI: 10.3389/fnmol.2022.1061343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Astrocytes (AST) are abundant glial cells in the human brain, accounting for approximately 20-50% percent of mammalian central nervous system (CNS) cells. They display essential functions necessary to sustain the physiological processes of the CNS, including maintaining neuronal structure, forming the blood-brain barrier, coordinating neuronal metabolism, maintaining the extracellular environment, regulating cerebral blood flow, stabilizing intercellular communication, participating in neurotransmitter synthesis, and defending against oxidative stress et al. During the pathological development of brain tumors, stroke, spinal cord injury (SCI), neurodegenerative diseases, and other neurological disorders, astrocytes undergo a series of highly heterogeneous changes, which are called reactive astrocytes, and mediate the corresponding pathophysiological process. However, the pathophysiological mechanisms of reactive astrocytes and their therapeutic relevance remain unclear. The microRNAs (miRNAs) are essential for cell differentiation, proliferation, and survival, which play a crucial role in the pathophysiological development of CNS diseases. In this review, we summarize the regulatory mechanism of miRNAs on reactive astrocytes in CNS diseases, which might provide a theoretical basis for the diagnosis and treatment of CNS diseases.
Collapse
|
22
|
Kurniati D, Hirai S, Egashira Y. Effect of apigenin on tryptophan metabolic key enzymes expression in lipopolysaccharide-induced microglial cells and its mechanism. Heliyon 2022; 9:e12743. [PMID: 36685364 PMCID: PMC9852672 DOI: 10.1016/j.heliyon.2022.e12743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/09/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
[Aims] Flavonoid apigenin (API) has a wide range of biological functions, particularly anti-inflammation. Indoleamine 2,3-dioxygenase (IDO) and 2-Amino-3-carboxymuconate-6-semialdehyde decarboxylase (ACMSD) are important tryptophan metabolic enzymes that play pivotal roles in the production of toxic metabolite quinolinic acid. However, the relationship between inflammation and ACMSD remains unclear. The present study investigated the relationship between inflammation and tryptophan metabolic key enzymes. Similarly, the anti-inflammatory effect of API on important tryptophan metabolic enzymes was examined in lipopolysaccharide (LPS)-treated microglial cells. [Main methods] MG6 cells were exposed to LPS with or without API treatment for 24-48 h. IDO and ACMSD mRNA expression and production of inflammatory mediators were analyzed. Activation of inflammatory signaling pathways, such as mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB), was also examined to study the mechanism of API in the inflammatory state. [Key findings] LPS suppressed ACMSD expression and enhanced IDO expression. However, API elevated ACMSD mRNA expression and suppressed IDO mRNA expression in LPS-treated MG6 cells. Furthermore, API suppressed interleukin-6 and nitric oxide production, whereas overproduction of inflammatory mediators enhanced IDO expression and assisted tryptophan degradation. API also inhibited activation of extracellular signal-regulated kinase (Erk) and jun N-terminal kinase (JNK) MAPK, and degradation of IκBα. [Significance] These results indicate alteration of ACMSD expression under inflammatory conditions. Moreover, API recovers expression of tryptophan metabolic key enzymes, which may be mediated by inhibition of proinflammatory mediator production via inactivation of Erk, JNK MAPK, and NF-κB pathways in LPS-stimulated microglial cells.
Collapse
Affiliation(s)
- Dian Kurniati
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan,Department of Food Technology, Faculty of Agricultural Industrial Technology, Universitas Padjadjaran, Sumedang KM. 21, Jatinangor, 40600, West Java, Indonesia
| | - Shizuka Hirai
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan
| | - Yukari Egashira
- Laboratory of Food and Nutrition, Division of Applied Biochemistry, Graduate School of Horticulture, Chiba University, 648, Matsudo, Matsudo-shi, Chiba, 271-8510, Japan,Corresponding author.
| |
Collapse
|
23
|
Basile MS, Mazzon E. The Role of Cannabinoid Type 2 Receptors in Parkinson's Disease. Biomedicines 2022; 10:biomedicines10112986. [PMID: 36428554 PMCID: PMC9687889 DOI: 10.3390/biomedicines10112986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disease and currently represents a clear unmet medical need. Therefore, novel preventive and therapeutic strategies are needed. Cannabinoid type 2 (CB2) receptors, one of the components of the endocannabinoid system, can regulate neuroinflammation in PD. Here, we review the current preclinical and clinical studies investigating the CB2 receptors in PD with the aim to clarify if these receptors could have a role in PD. Preclinical data show that CB2 receptors could have a neuroprotective action in PD and that the therapeutic targeting of CB2 receptors could be promising. Indeed, it has been shown that different CB2 receptor-selective agonists exert protective effects in different PD models. Moreover, the alterations in the expression of CB2 receptors observed in brain tissues from PD animal models and PD patients suggest the potential value of CB2 receptors as possible novel biomarkers for PD. However, to date, there is no direct evidence of the role of CB2 receptors in PD. Further studies are strongly needed in order to fully clarify the role of CB2 receptors in PD and thus pave the way to novel possible diagnostic and therapeutic opportunities for PD.
Collapse
Affiliation(s)
- Maria Sofia Basile
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
24
|
Wang W, Jiang S, Xu C, Tang L, Liang Y, Zhao Y, Zhu G. Interactions between gut microbiota and Parkinson's disease: The role of microbiota-derived amino acid metabolism. Front Aging Neurosci 2022; 14:976316. [PMID: 36408101 PMCID: PMC9667037 DOI: 10.3389/fnagi.2022.976316] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/29/2022] [Indexed: 11/05/2022] Open
Abstract
Non-motor symptoms (NMS) of Parkinson's disease (PD), such as constipation, sleep disorders, and olfactory deficits, may emerge up to 20 years earlier than motor symptoms. A series of evidence indicates that the pathology of PD may occur from the gastrointestinal tract to the brain. Numerous studies support that the gut microbiota communicates with the brain through the immune system, special amino acid metabolism, and the nervous system in PD. Recently, there is growing recognition that the gut microbiota plays a vital role in the modulation of multiple neurochemical pathways via the “gut microbiota-brain axis” (GMBA). Many gut microbiota metabolites, such as fatty acids, amino acids, and bile acids, convey signaling functions as they mediate the crosstalk between gut microbiota and host physiology. Amino acids' abundance and species alteration, including glutamate and tryptophan, may disturb the signaling transmission between nerve cells and disrupt the normal basal ganglia function in PD. Specific amino acids and their receptors are considered new potential targets for ameliorating PD. The present study aimed to systematically summarize all available evidence on the gut microbiota-derived amino acid metabolism alterations associated with PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujun Jiang
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengcheng Xu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Tang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Yang Zhao
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Guoxue Zhu
| |
Collapse
|
25
|
Bhardwaj A, Bhardwaj R, Saini A, Dhawan DK, Kaur T. Impact of Calcium Influx on Endoplasmic Reticulum in Excitotoxic Neurons: Role of Chemical Chaperone 4-PBA. Cell Mol Neurobiol 2022; 43:1619-1635. [PMID: 36002608 DOI: 10.1007/s10571-022-01271-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
Abstract
Excessive activation of α-amino-3-hydroxy-5-methyl-4-isoxazole propoinic acid (AMPA) receptors instigates excitotoxicity via enhanced calcium influx in the neurons thus inciting deleterious consequences. Additionally, Endoplasmic Reticulum (ER) is pivotal in maintaining the intracellular calcium balance. Considering this, studying the aftermath of enhanced calcium uptake by neurons and its effect on ER environment can assist in delineating the pathophysiological events incurred by excitotoxicty. The current study was premeditated to decipher the role of ER pertaining to calcium homeostasis in AMPA-induced excitotoxicity. The findings showed, increased intracellular calcium levels (measured by flowcytometry and spectroflourimeter using Fura 2AM) in AMPA excitotoxic animals (male Sprague dawely rats) (intra-hippocampal injection of 10 mM AMPA). Further, ER resident proteins like calnexin, PDI and ERp72 were found to be upregulated, which further modulated the functioning of ER membrane calcium channels viz. IP3R, RyR, and SERCA pump. Altered calcium homeostasis further led to ER stress and deranged the protein folding capacity of ER post AMPA toxicity, which was ascertained by unfolded protein response (UPR) pathway markers such as IRE1α, eIF2α, and ATF6α. Chemical chaperone, 4-phenybutric acid (4-PBA), ameliorated the protein folding capacity and subsequent UPR markers. In addition, modulation of calcium channels and calcium regulating machinery of ER post 4-PBA administration restored the calcium homeostasis. Therefore the study reinforces the significance of ER stress, a debilitating outcome of impaired calcium homeostasis, under AMPA-induced excitotoxicity. Also, employing chaperone-based therapeutic approach to curb ER stress can restore the calcium imbalance in the neuropathological diseases.
Collapse
Affiliation(s)
- Ankita Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Rishi Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | | | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
26
|
Gold OMS, Bardsley EN, Ponnampalam AP, Pauza AG, Paton JFR. Cellular basis of learning and memory in the carotid body. Front Synaptic Neurosci 2022; 14:902319. [PMID: 36046221 PMCID: PMC9420943 DOI: 10.3389/fnsyn.2022.902319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The carotid body is the primary peripheral chemoreceptor in the body, and critical for respiration and cardiovascular adjustments during hypoxia. Yet considerable evidence now implicates the carotid body as a multimodal sensor, mediating the chemoreflexes of a wide range of physiological responses, including pH, temperature, and acidosis as well as hormonal, glucose and immune regulation. How does the carotid body detect and initiate appropriate physiological responses for these diverse stimuli? The answer to this may lie in the structure of the carotid body itself. We suggest that at an organ-level the carotid body is comparable to a miniature brain with compartmentalized discrete regions of clustered glomus cells defined by their neurotransmitter expression and receptor profiles, and with connectivity to defined reflex arcs that play a key role in initiating distinct physiological responses, similar in many ways to a switchboard that connects specific inputs to selective outputs. Similarly, within the central nervous system, specific physiological outcomes are co-ordinated, through signaling via distinct neuronal connectivity. As with the brain, we propose that highly organized cellular connectivity is critical for mediating co-ordinated outputs from the carotid body to a given stimulus. Moreover, it appears that the rudimentary components for synaptic plasticity, and learning and memory are conserved in the carotid body including the presence of glutamate and GABAergic systems, where evidence pinpoints that pathophysiology of common diseases of the carotid body may be linked to deviations in these processes. Several decades of research have contributed to our understanding of the central nervous system in health and disease, and we discuss that understanding the key processes involved in neuronal dysfunction and synaptic activity may be translated to the carotid body, offering new insights and avenues for therapeutic innovation.
Collapse
|
27
|
Zhang C, Ni C, Lu H. Polo-Like Kinase 2: From Principle to Practice. Front Oncol 2022; 12:956225. [PMID: 35898867 PMCID: PMC9309260 DOI: 10.3389/fonc.2022.956225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/21/2022] Open
Abstract
Polo-like kinase (PLK) 2 is an evolutionarily conserved serine/threonine kinase that shares the n-terminal kinase catalytic domain and the C-terminal Polo Box Domain (PBD) with other members of the PLKs family. In the last two decades, mounting studies have focused on this and tried to clarify its role in many aspects. PLK2 is essential for mitotic centriole replication and meiotic chromatin pairing, synapsis, and crossing-over in the cell cycle; Loss of PLK2 function results in cell cycle disorders and developmental retardation. PLK2 is also involved in regulating cell differentiation and maintaining neural homeostasis. In the process of various stimuli-induced stress, including oxidative and endoplasmic reticulum, PLK2 may promote survival or apoptosis depending on the intensity of stimulation and the degree of cell damage. However, the role of PLK2 in immunity to viral infection has been studied far less than that of other family members. Because PLK2 is extensively and deeply involved in normal physiological functions and pathophysiological mechanisms of cells, its role in diseases is increasingly being paid attention to. The effect of PLK2 in inhibiting hematological tumors and fibrotic diseases, as well as participating in neurodegenerative diseases, has been gradually recognized. However, the research results in solid organ tumors show contradictory results. In addition, preliminary studies using PLK2 as a disease predictor and therapeutic target have yielded some exciting and promising results. More research will help people better understand PLK2 from principle to practice.
Collapse
Affiliation(s)
- Chuanyong Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Chuangye Ni
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Hao Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
- *Correspondence: Hao Lu,
| |
Collapse
|
28
|
GLP-1 Receptor Agonists in Neurodegeneration: Neurovascular Unit in the Spotlight. Cells 2022; 11:cells11132023. [PMID: 35805109 PMCID: PMC9265397 DOI: 10.3390/cells11132023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Defects in brain energy metabolism and proteopathic stress are implicated in age-related degenerative neuronopathies, exemplified by Alzheimer’s disease (AD) and Parkinson’s disease (PD). As the currently available drug regimens largely aim to mitigate cognitive decline and/or motor symptoms, there is a dire need for mechanism-based therapies that can be used to improve neuronal function and potentially slow down the underlying disease processes. In this context, a new class of pharmacological agents that achieve improved glycaemic control via the glucagon-like peptide 1 (GLP-1) receptor has attracted significant attention as putative neuroprotective agents. The experimental evidence supporting their potential therapeutic value, mainly derived from cellular and animal models of AD and PD, has been discussed in several research reports and review opinions recently. In this review article, we discuss the pathological relevance of derangements in the neurovascular unit and the significance of neuron–glia metabolic coupling in AD and PD. With this context, we also discuss some unresolved questions with regard to the potential benefits of GLP-1 agonists on the neurovascular unit (NVU), and provide examples of novel experimental paradigms that could be useful in improving our understanding regarding the neuroprotective mode of action associated with these agents.
Collapse
|
29
|
Oikawa T, Fukuda T, Yamashita T, Tomita H, Ozaki T. Lentiviral expression of calpain-1 C2-like domain peptide prevents glutamate-induced cell death in mouse hippocampal neuronal HT22 cells. In Vitro Cell Dev Biol Anim 2022; 58:289-294. [PMID: 35469046 DOI: 10.1007/s11626-022-00683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/30/2022] [Indexed: 11/05/2022]
Abstract
Glutamate neurotoxicity is involved in neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. Excess glutamate causes caspase-independent programmed cell death via oxidative stress and calcium influx. Our previous study showed that calpain-1 localizes to both the cytoplasm and mitochondria, where apoptosis-inducing factor (AIF) is cleaved by calpain-1 and translocates to the nucleus to induce DNA fragmentation. The autoinhibitory region of calpain-1 conjugated with the cell-penetrating peptide HIV1-Tat (namely Tat-μCL) specifically prevents the activity of mitochondrial calpain-1 and attenuates neuronal cell death in animal models of retinitis pigmentosa, as well as glutamate-induced cell death in mouse hippocampal HT22 cells. In the present study, we constructed a lentiviral vector expressing the Tat-μCL peptide and evaluated its protective effect against glutamate-induced cell death in HT22 cells. Lentiviral transduction with Tat-μCL significantly suppressed glutamate-induced nuclear translocation of AIF and DNA fragmentation. The findings of the present study suggest that the stable expression of Tat-μCL may be a potential gene therapy modality for neurodegenerative diseases.
Collapse
Affiliation(s)
- Takenori Oikawa
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Tomokazu Fukuda
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate, 020-8550, Japan
| | - Hiroshi Tomita
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Taku Ozaki
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| |
Collapse
|
30
|
Hazarika I, Mukundan GK, Sundari PS. Neuroprotective effect of Hydrocotyle sibthorpioides against monosodium glutamate-induced excitotoxicity. Nat Prod Res 2022; 36:6156-6159. [PMID: 35356842 DOI: 10.1080/14786419.2022.2057493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We aimed to evaluate the neuroprotective effect of H. sibthorpioides against monosodium-glutamate (MSG) induced excitoneurotoxicity in rats. We randomly divided the animals into 11 groups (n = 8) and subjected them to high doses of MSG (2 g/kg body weight) and the test dose (1 week). The test chemicals were H. sibthorpioides extracts of petroleum ether, chloroform, methanol, and water. We used Dizocilpine-hydrogen-maleate as a standard and assessed the cognitive property using Morris-water-maze and elevated-plus-maze. After the experimental period, we evaluated the biochemical parameters. We found chloroform and methanolic extracts significantly enhanced the cognitive behaviour of rats compared to control. Biochemical analysis suggested that there was a high level of antioxidants and lower levels of glutamate and proinflammatory cytokines in the cortex and hippocampus. We concluded that chloroform and methanolic extracts of H. sibthorpioides enhanced the level of antioxidants, decreased proinflammatory-cytokines and glutamate in the brain, and thus prevented the monosodium-glutamate-induced-excite-neurotoxicity.
Collapse
Affiliation(s)
- Iswar Hazarika
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka, India.,Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science, Guwahati, Assam, India
| | - Geetha Kannoth Mukundan
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka, India
| | - P Sivakami Sundari
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka, India
| |
Collapse
|
31
|
Striatal glutamatergic hyperactivity in Parkinson's disease. Neurobiol Dis 2022; 168:105697. [DOI: 10.1016/j.nbd.2022.105697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
|
32
|
Luttenbacher I, Phillips A, Kazemi R, Hadipour AL, Sanghvi I, Martinez J, Adamson MM. Transdiagnostic role of glutamate and white matter damage in neuropsychiatric disorders: A Systematic Review. J Psychiatr Res 2022; 147:324-348. [PMID: 35151030 DOI: 10.1016/j.jpsychires.2021.12.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 12/09/2022]
Abstract
Neuropsychiatric disorders including generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SZ) have been considered distinct categories of diseases despite their overlapping characteristics and symptomatology. We aimed to provide an in-depth review elucidating the role of glutamate/Glx and white matter (WM) abnormalities in these disorders from a transdiagnostic perspective. The PubMed online database was searched for studies published between 2010 and 2021. After careful screening, 401 studies were included. The findings point to decreased levels of glutamate in the Anterior Cingulate Cortex in both SZ and BD, whereas Glx is elevated in the Hippocampus in SZ and MDD. With regard to WM abnormalities, the Corpus Callosum and superior Longitudinal Fascicle were the most consistently identified brain regions showing decreased fractional anisotropy (FA) across all the reviewed disorders, except GAD. Additionally, the Uncinate Fasciculus displayed decreased FA in all disorders, except OCD. Decreased FA was also found in the inferior Longitudinal Fasciculus, inferior Fronto-Occipital Fasciculus, Thalamic Radiation, and Corona Radiata in SZ, BD, and MDD. Decreased FA in the Fornix and Corticospinal Tract were found in BD and SZ patients. The Cingulum and Anterior Limb of Internal Capsule exhibited decreased FA in MDD and SZ patients. The results suggest a gradual increase in severity from GAD to SZ defined by the number of brain regions with WM abnormality which may be partially caused by abnormal glutamate levels. WM damage could thus be considered a potential marker of some of the main neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ines Luttenbacher
- Department of Social & Behavioral Sciences, University of Amsterdam, Amsterdam, Netherlands; Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Angela Phillips
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Reza Kazemi
- Department of Cognitive Psychology, Institute for Cognitive Science Studies, Tehran, Iran
| | - Abed L Hadipour
- Department of Cognitive Sciences, University of Messina, Messina, Italy
| | - Isha Sanghvi
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neuroscience, University of Southern California, Los Angeles, CA, USA
| | - Julian Martinez
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Palo Alto University, Palo Alto, CA, USA
| | - Maheen M Adamson
- Rehabilitation Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
33
|
Wang M, Liu H, Ma Z. Roles of the Cannabinoid System in the Basal Ganglia in Parkinson’s Disease. Front Cell Neurosci 2022; 16:832854. [PMID: 35264932 PMCID: PMC8900732 DOI: 10.3389/fncel.2022.832854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/31/2022] [Indexed: 12/26/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease usually caused by neuroinflammation, oxidative stress and other etiologies. Recent studies have found that the cannabinoid system present in the basal ganglia has a strong influence on the progression of PD. Altering the cannabinoid receptor activation status by modulating endogenous cannabinoid (eCB) levels can exert an anti-movement disorder effect. Therefore, the development of drugs that modulate the endocannabinoid system may be a novel strategy for the treatment of PD. However, eCB regulation is complex, with diverse cannabinoid receptor functions and the presence of dopaminergic, glutamatergic, and γ-aminobutyric signals interacting with cannabinoid signaling in the basal ganglia region. Therefore, the study of eCB is challenging. Here, we have described the function of the cannabinoid system in the basal ganglia and its association with PD in three parts (eCBs, cannabinoid receptors, and factors regulating the cannabinoid metabolism) and summarized the mechanisms of action related to the cannabinoid analogs currently aimed at treating PD. The shortcomings identified from previous studies and the directions that should be explored in the future will provide insights into new approaches and ideas for the future development of cannabinoid-based drugs and the treatment of PD.
Collapse
Affiliation(s)
- Mengya Wang
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, China
| | - Huayuan Liu
- Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Zegang Ma
- Department of Physiology, School of Basic Medicine, Institute of Brain Science and Disorders, Qingdao University, Qingdao, China
- *Correspondence: Zegang Ma,
| |
Collapse
|
34
|
Bancroft EA, Srinivasan R. Emerging Roles for Aberrant Astrocytic Calcium Signals in Parkinson's Disease. Front Physiol 2022; 12:812212. [PMID: 35087422 PMCID: PMC8787054 DOI: 10.3389/fphys.2021.812212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/09/2021] [Indexed: 11/25/2022] Open
Abstract
Astrocytes display a plethora of spontaneous Ca2+ signals that modulate vital functions of the central nervous system (CNS). This suggests that astrocytic Ca2+ signals also contribute to pathological processes in the CNS. In this context, the molecular mechanisms by which aberrant astrocytic Ca2+ signals trigger dopaminergic neuron loss during Parkinson's disease (PD) are only beginning to emerge. Here, we provide an evidence-based perspective on potential mechanisms by which aberrant astrocytic Ca2+ signals can trigger dysfunction in three distinct compartments of the brain, viz., neurons, microglia, and the blood brain barrier, thereby leading to PD. We envision that the coming decades will unravel novel mechanisms by which aberrant astrocytic Ca2+ signals contribute to PD and other neurodegenerative processes in the CNS.
Collapse
Affiliation(s)
- Eric A. Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Rahul Srinivasan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University College of Medicine, Bryan, TX, United States
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, United States
| |
Collapse
|
35
|
NMDA mediates disruption of blood-brain barrier permeability via Rho/ROCK signaling pathway. Neurochem Int 2022; 154:105278. [PMID: 35017026 DOI: 10.1016/j.neuint.2022.105278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/21/2021] [Accepted: 01/05/2022] [Indexed: 01/14/2023]
Abstract
Glutamate can activate the N-methyl-D-aspartatic acid (NMDA) receptor (NMDAR), damage brain microvascular endothelial cells, and disturb the intercellular tight junctions (TJs). These result in changes in the permeability of the blood brain barrier (BBB). In neurons, the activation of Rho/ROCK signaling pathway is related to the activation of NMDAR,however, whether human brain vascular endothelial cells NMDAR mediates the Rho/ROCK pathway is not fully understood. The present study evaluates the effects of excessive NMDAR activation induced by NMDA (a glutamate analog) on the Rho/ROCK signaling pathway and the permeability of BBB by using a primary human brain microvascular endothelial cell (HBMEC) model. NMDAR subunit GluN1 was expressed in HBMECs and promoted by NMDA detected by Western blot and qRT-PCR. Furthermore, NMDA exposure decreased HBMEC viability, promoted HBMEC apoptosis, increased intracellular reactive oxygen species (ROS) levels, and destroyed the endothelial cytoskeleton. Additionally, NMDA exposure suppressed transendothelial electrical resistance (TEER) values and the expression of TJ proteins occludin and claudin5; it also promoted ROCK activated substrate myosin phosphatase target subunit-1 (MYPT)-1 phosphorylation and the transmittance of sodium fluorescein. In contrast, these effects were attenuated by ROCK inhibitor hydroxyfasudil (HF) and NMDAR antagonist MK801, respectively. Therefore, these results indicate that excessive endothelial NMDAR activation induced by NMDA may induce TJs and cytoskeleton damage, while HF attenuated NMDA-induced cytotoxicity in HBMECs by inhibiting the Rho/ROCK signaling pathway.
Collapse
|
36
|
Bentea E, De Pauw L, Verbruggen L, Winfrey LC, Deneyer L, Moore C, Albertini G, Sato H, Van Eeckhaut A, Meshul CK, Massie A. Aged xCT-Deficient Mice Are Less Susceptible for Lactacystin-, but Not 1-Methyl-4-Phenyl-1,2,3,6- Tetrahydropyridine-, Induced Degeneration of the Nigrostriatal Pathway. Front Cell Neurosci 2022; 15:796635. [PMID: 34975413 PMCID: PMC8718610 DOI: 10.3389/fncel.2021.796635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
The astrocytic cystine/glutamate antiporter system x c - (with xCT as the specific subunit) imports cystine in exchange for glutamate and has been shown to interact with multiple pathways in the brain that are dysregulated in age-related neurological disorders, including glutamate homeostasis, redox balance, and neuroinflammation. In the current study, we investigated the effect of genetic xCT deletion on lactacystin (LAC)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced degeneration of the nigrostriatal pathway, as models for Parkinson's disease (PD). Dopaminergic neurons of adult xCT knock-out mice (xCT-/-) demonstrated an equal susceptibility to intranigral injection of the proteasome inhibitor LAC, as their wild-type (xCT+/+) littermates. Contrary to adult mice, aged xCT-/- mice showed a significant decrease in LAC-induced degeneration of nigral dopaminergic neurons, depletion of striatal dopamine (DA) and neuroinflammatory reaction, compared to age-matched xCT+/+ littermates. Given this age-related protection, we further investigated the sensitivity of aged xCT-/- mice to chronic and progressive MPTP treatment. However, in accordance with our previous observations in adult mice (Bentea et al., 2015a), xCT deletion did not confer protection against MPTP-induced nigrostriatal degeneration in aged mice. We observed an increased loss of nigral dopaminergic neurons, but equal striatal DA denervation, in MPTP-treated aged xCT-/- mice when compared to age-matched xCT+/+ littermates. To conclude, we reveal age-related protection against proteasome inhibition-induced nigrostriatal degeneration in xCT-/- mice, while xCT deletion failed to protect nigral dopaminergic neurons of aged mice against MPTP-induced toxicity. Our findings thereby provide new insights into the role of system x c - in mechanisms of dopaminergic cell loss and its interaction with aging.
Collapse
Affiliation(s)
- Eduard Bentea
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lise Verbruggen
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lila C Winfrey
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Lauren Deneyer
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cynthia Moore
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Giulia Albertini
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata, Japan
| | - Ann Van Eeckhaut
- Research Group Experimental Pharmacology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | - Charles K Meshul
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States.,Department of Behavioral Neuroscience and Pathology, Oregon Health and Science University, Portland, OR, United States
| | - Ann Massie
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
37
|
Neuroprotective Effect of Ceftriaxone on MPTP-Induced Parkinson's Disease Mouse Model by Regulating Inflammation and Intestinal Microbiota. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9424582. [PMID: 34938384 PMCID: PMC8687851 DOI: 10.1155/2021/9424582] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/20/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023]
Abstract
Parkinson's disease (PD) is a common degenerative disease of the central nervous system. Although some drugs can alleviate the progress of PD, their long-term use will lead to complications, so it is still necessary to find new drugs to delay or cure PD effectively. In view of the difficulty in developing new drugs, it is imperative to discover new functions of existing compounds that could be used to treat PD. In this study, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was used to induce PD symptoms in a mouse model. Subsequently, these mice were treated with the antibiotic ceftriaxone. Ceftriaxone alleviated the behavioural and neuropathological changes induced by MPTP, downregulated the expression of glial fibrillary acidic protein (GFAP) and ionised calcium-binding adapter molecule 1 (Iba1) as markers of astroglia and microglia, respectively, and reduced the expression of neuroinflammation-related Toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and phosphorylated nuclear factor kappa-B (p-NF-κB)/NF-κB in the brain of PD mice. In addition, ceftriaxone reduced the abundance of pathogenic bacteria of the genus Proteus and increased the abundance of probiotic Akkermansia. Finally, ceftriaxone treatment increased the expression of the tight junction proteins zona occludens-1(ZO-1) and occludin in the colon, decreased the expression of the inflammation-related proteins TLR4, MyD88, and NF-κB in the colon, and decreased the serum concentration of the proinflammatory cytokines interleukin-1β (IL-1β), IL-6, and tumour necrosis factor-α (TNF-α). These results indicate that ceftriaxone had a neuroprotective effect on MPTP-induced PD mice, and its neuroprotective effect could be through regulating inflammation and intestinal microbiota. While we showed that ceftriaxone exerts a neuroprotective effect in an MPTP-induced PD mouse model, our findings are limited to the short-term effects of ceftriaxone. Additional work using transgenic mice is required to determine the long-term effects of ceftriaxone. In addition, the dose and frequency of ceftriaxone use should be evaluated.
Collapse
|
38
|
Knorr S, Musacchio T, Paulat R, Matthies C, Endres H, Wenger N, Harms C, Ip CW. Experimental deep brain stimulation in rodent models of movement disorders. Exp Neurol 2021; 348:113926. [PMID: 34793784 DOI: 10.1016/j.expneurol.2021.113926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/14/2021] [Accepted: 11/11/2021] [Indexed: 12/21/2022]
Abstract
Deep brain stimulation (DBS) is the preferred treatment for therapy-resistant movement disorders such as dystonia and Parkinson's disease (PD), mostly in advanced disease stages. Although DBS is already in clinical use for ~30 years and has improved patients' quality of life dramatically, there is still limited understanding of the underlying mechanisms of action. Rodent models of PD and dystonia are essential tools to elucidate the mode of action of DBS on behavioral and multiscale neurobiological levels. Advances have been made in identifying DBS effects on the central motor network, neuroprotection and neuroinflammation in DBS studies of PD rodent models. The phenotypic dtsz mutant hamster and the transgenic DYT-TOR1A (ΔETorA) rat proved as valuable models of dystonia for preclinical DBS research. In addition, continuous refinements of rodent DBS technologies are ongoing and have contributed to improvement of experimental quality. We here review the currently existing literature on experimental DBS in PD and dystonia models regarding the choice of models, experimental design, neurobiological readouts, as well as methodological implications. Moreover, we provide an overview of the technical stage of existing DBS devices for use in rodent studies.
Collapse
Affiliation(s)
- Susanne Knorr
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, Würzburg, Germany.
| | - Thomas Musacchio
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, Würzburg, Germany.
| | - Raik Paulat
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, Germany.
| | - Cordula Matthies
- Department of Neurosurgery, University Hospital of Würzburg, Josef-Schneider-Straße 11, Würzburg, Germany.
| | - Heinz Endres
- University of Applied Science Würzburg-Schweinfurt, Schweinfurt, Germany.
| | - Nikolaus Wenger
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, Germany.
| | - Christoph Harms
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, Germany.
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, Würzburg, Germany.
| |
Collapse
|
39
|
Ren C, He KJ, Hu H, Zhang JB, Dong LG, Li D, Chen J, Mao CJ, Wang F, Liu CF. Induction of Parkinsonian-Like Changes via Targeted Downregulation of Astrocytic Glutamate Transporter GLT-1 in the Striatum. JOURNAL OF PARKINSONS DISEASE 2021; 12:295-314. [PMID: 34719508 DOI: 10.3233/jpd-212640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous investigations have suggested that decreased expression of glutamate transporter-1 (GLT-1) is involved in glutamate excitotoxicity and contribute to the development of Parkinson's disease (PD), GLT-1 is decreased in animal models of PD. GLT-1 is mainly expressed in astrocytes, and the striatum is a GLT-1-rich brain area. OBJECTIVE The aim was to explore the function and mechanism of astrocytic GLT-1 in PD-like changes. METHODS In the study, PD-like changes and their molecular mechanism in rodents were tested by a behavioral assessment, micro-positron emission tomography/computed tomography (PET/CT), western blotting, immunohistochemical and immunofluorescence staining, and high performance liquid chromatography pre-column derivatization with O-pthaldialdehida after downregulating astrocytic GLT-1 in vivo and in vitro. RESULTS In vivo, after 6 weeks of brain stereotactic injection of adeno-associated virus into the striatum, rats in the astrocytic GLT-1 knockdown group showed poorer motor performance, abnormal gait, and depression-like feature; but no olfactory disorders. The results of micro-PET/CT and western blotting indicated that the dopaminergic system was impaired in astrocytic GLT-1 knockdown rats. Similarly, tyrosine hydroxylase (TH) positive immune-staining in neurons of astrocytic GLT-1 knockdown rats showed deficit in cell count. In vitro, knockdown of astrocytic GLT-1 via RNA interference led to morphological injury of TH-positive neurons, which may be related to the abnormal calcium signal induced by glutamate accumulation after GLT-1 knockdown. Furthermore, the GLT-1 agonist ceftriaxone showed a protective effect on TH-positive neuron impairment. CONCLUSION The present findings may shed new light on the future prevention and treatment of PD based on blocking glutamate excitotoxicity.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kai-Jie He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hua Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dan Li
- Department of Neurology, Suqian First Hospital, Suqian, China
| | - Jing Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, Suqian First Hospital, Suqian, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China.,Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
40
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
41
|
A nanophytosomes formulation based on elderberry anthocyanins and Codium lipids to mitigate mitochondrial dysfunctions. Biomed Pharmacother 2021; 143:112157. [PMID: 34517282 DOI: 10.1016/j.biopha.2021.112157] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
The development of nanomedicines to modulate the mitochondrial function is a great scientific challenge since mitochondrial dysfunction is a pathological hallmark of many chronic diseases, including degenerative brain pathologies like Parkinson's and Alzheimer's diseases. To address this challenge, the mitochondriotropic features of the elderberry anthocyanin-enriched extract (Sambucus nigra) were combined with the self-assembling properties of the membrane polar lipids from Codium tomentosum in an innovative SC-Nanophytosomes formulation. Membrane polar lipids, obtained by a new procedure as chlorophyll-free extract, are characterized by 26% of non-phosphorus polar lipids and 74% of phospholipids (dominated by anionic lipids) containing a high degree of polyunsaturated fatty acids. The anthocyanin-enriched extract is dominated by a mixture of four cyanidin-glycosides, representing about 86% of their phenolic content. SC-Nanophytosomes engineered with 600 µM algae membrane polar lipids and 0.5 mg/L of the anthocyanin-enriched extract are nanosized vesicles (diameter =108.74 ± 24.74 nm) with a negative surface charge (Zeta potential = -46.93 ± 6.63 mV) that exhibit stability during storage at 4 ºC. In vitro assays with SH-SY5Y cells showed that SC-Nanophytosomes have the competence to target mitochondria, improving the mitochondrial respiratory chain complexes I and II and preserving the mitochondrial membrane potential in the presence of rotenone. Additionally, SC-Nanophytosomes protect SH-SY5Y cells against the toxicity induced by rotenone or glutamate. Green-fluorescent labeled SC-Nanophytosomes were used to reveal that they are mainly internalized by cells via caveola-mediated endocytosis, escape from endosome and reach the cytoplasm organelles, including mitochondria. Overall, data indicate that SC-Nanophytosomes have the potential to support a mitochondria-targeted therapy for neurodegenerative diseases.
Collapse
|
42
|
Guo C, Ma YY. Calcium Permeable-AMPA Receptors and Excitotoxicity in Neurological Disorders. Front Neural Circuits 2021; 15:711564. [PMID: 34483848 PMCID: PMC8416103 DOI: 10.3389/fncir.2021.711564] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Excitotoxicity is one of the primary mechanisms of cell loss in a variety of diseases of the central and peripheral nervous systems. Other than the previously established signaling pathways of excitotoxicity, which depend on the excessive release of glutamate from axon terminals or over-activation of NMDA receptors (NMDARs), Ca2+ influx-triggered excitotoxicity through Ca2+-permeable (CP)-AMPA receptors (AMPARs) is detected in multiple disease models. In this review, both acute brain insults (e.g., brain trauma or spinal cord injury, ischemia) and chronic neurological disorders, including Epilepsy/Seizures, Huntington’s disease (HD), Parkinson’s disease (PD), Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), chronic pain, and glaucoma, are discussed regarding the CP-AMPAR-mediated excitotoxicity. Considering the low expression or absence of CP-AMPARs in most cells, specific manipulation of the CP-AMPARs might be a more plausible strategy to delay the onset and progression of pathological alterations with fewer side effects than blocking NMDARs.
Collapse
Affiliation(s)
- Changyong Guo
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yao-Ying Ma
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
43
|
Guglietti B, Sivasankar S, Mustafa S, Corrigan F, Collins-Praino LE. Fyn Kinase Activity and Its Role in Neurodegenerative Disease Pathology: a Potential Universal Target? Mol Neurobiol 2021; 58:5986-6005. [PMID: 34432266 DOI: 10.1007/s12035-021-02518-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Fyn is a non-receptor tyrosine kinase belonging to the Src family of kinases (SFKs) which has been implicated in several integral functions throughout the central nervous system (CNS), including myelination and synaptic transmission. More recently, Fyn dysfunction has been associated with pathological processes observed in neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). Neurodegenerative diseases are amongst the leading cause of death and disability worldwide and, due to the ageing population, prevalence is predicted to rise in the coming years. Symptoms across neurodegenerative diseases are both debilitating and degenerative in nature and, concerningly, there are currently no disease-modifying therapies to prevent their progression. As such, it is important to identify potential new therapeutic targets. This review will outline the role of Fyn in normal/homeostatic processes, as well as degenerative/pathological mechanisms associated with neurodegenerative diseases, such as demyelination, pathological protein aggregation, neuroinflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Bianca Guglietti
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Srisankavi Sivasankar
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Sanam Mustafa
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia
| | - Frances Corrigan
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia. .,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
44
|
Thomas GEC, Zarkali A, Ryten M, Shmueli K, Gil-Martinez AL, Leyland LA, McColgan P, Acosta-Cabronero J, Lees AJ, Weil RS. Regional brain iron and gene expression provide insights into neurodegeneration in Parkinson's disease. Brain 2021; 144:1787-1798. [PMID: 33704443 PMCID: PMC8320305 DOI: 10.1093/brain/awab084] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/20/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanisms responsible for the selective vulnerability of specific neuronal populations in Parkinson's disease are poorly understood. Oxidative stress secondary to brain iron accumulation is one postulated mechanism. We measured iron deposition in 180 cortical regions of 96 patients with Parkinson's disease and 35 control subjects using quantitative susceptibility mapping. We estimated the expression of 15 745 genes in the same regions using transcriptomic data from the Allen Human Brain Atlas. Using partial least squares regression, we then identified the profile of gene transcription in the healthy brain that underlies increased cortical iron in patients with Parkinson's disease relative to controls. Applying gene ontological tools, we investigated the biological processes and cell types associated with this transcriptomic profile and identified the sets of genes with spatial expression profiles in control brains that correlated significantly with the spatial pattern of cortical iron deposition in Parkinson's disease. Gene ontological analyses revealed that these genes were enriched for biological processes relating to heavy metal detoxification, synaptic function and nervous system development and were predominantly expressed in astrocytes and glutamatergic neurons. Furthermore, we demonstrated that the genes differentially expressed in Parkinson's disease are associated with the pattern of cortical expression identified in this study. Our findings provide mechanistic insights into regional selective vulnerabilities in Parkinson's disease, particularly the processes involving iron accumulation.
Collapse
Affiliation(s)
| | | | - Mina Ryten
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1B 5EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL, London, WC1N 1EH, UK
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | - Karin Shmueli
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, UCL, London, WC1E 6BT, UK
| | - Ana Luisa Gil-Martinez
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1B 5EH, UK
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | | | - Peter McColgan
- Huntington’s Disease Centre, UCL Institute of Neurology, London, WC1B 5EH, UK
| | | | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies, London, WC1N 1PJ, UK
| | - Rimona S Weil
- Dementia Research Centre, UCL, London, WC1N 3AR, UK
- Wellcome Centre for Human Neuroimaging, UCL, London, WC1N 3AR, UK
- Movement Disorders Consortium, UCL, London, WC1N 3BG, UK
| |
Collapse
|
45
|
Máñez-Miró JU, Rodríguez-Rojas R, Del Álamo M, Martínez-Fernández R, Obeso JA. Present and future of subthalamotomy in the management of Parkinson´s disease: a systematic review. Expert Rev Neurother 2021; 21:533-545. [PMID: 33788645 DOI: 10.1080/14737175.2021.1911649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: The subthalamic nucleus (STN) is known to be involved in the pathophysiology of Parkinson´s disease and by reducing its abnormal activity, normal output of basal ganglia can be restored along with improvement in PD cardinal motor features. Deep brain stimulation of the STN is currently the main surgical procedure for PD with motor complications, but lesioning can be an alternative.Areas covered: Here, the authors systematically review the current evidence regarding subthalamotomy both with radiofrequency and, more recently, with focused ultrasound (FUS) for the treatment of PD.Expert opinion: Unilateral subthalamotomy for the treatment of PD motor features can be considered a viable option in asymmetric patients, particularly with FUS which allows a minimally invasive safe and effective ablation of the STN. Risk of inducing dyskinesia (i.e., hemichorea/ballism) may be strikingly reduced when lesions enlarge dorsally to impinge on pallidothalamic fibers.
Collapse
Affiliation(s)
- Jorge U Máñez-Miró
- HM CINAC (Centro Integral De Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Rafael Rodríguez-Rojas
- HM CINAC (Centro Integral De Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Marta Del Álamo
- HM CINAC (Centro Integral De Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - R Martínez-Fernández
- HM CINAC (Centro Integral De Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - José A Obeso
- HM CINAC (Centro Integral De Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain.,CEU-San Pablo University, Móstoles, Madrid, Spain
| |
Collapse
|
46
|
Meng X, Zhong J, Zeng C, Yung KKL, Zhang X, Wu X, Qu S. MiR-30a-5p Regulates GLT-1 Function via a PKCα-Mediated Ubiquitin Degradation Pathway in a Mouse Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:1578-1592. [PMID: 33882234 DOI: 10.1021/acschemneuro.1c00076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Glutamate excitotoxicity is caused by dysfunctional glutamate transporters and plays an important role in the pathogenesis of Parkinson's disease (PD); however, the mechanisms that underlie the regulation of glutamate transporters in PD are still not fully elucidated. MicroRNAs(miRNA), which are abundant in astrocytes and neurons, have been reported to play key roles in regulating the translation of glutamate-transporter mRNA. In this study, we hypothesized that the miR-30a-5p contributes to the pathogenesis of PD by regulating the ubiquitin-mediated degradation of glutamate transporter 1 (GLT-1). We demonstrated that short-hairpin RNA-mediated knockdown of miR-30a-5p ameliorated motor deficits and pathological changes like astrogliosis and reactive microgliosis in a mouse model of PD (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice). Western blotting and immunofluorescent labeling revealed that miR-30a-5p suppressed the expression and function of GLT-1 in MPTP-treated mice and specifically in astrocytes treated with 1-methyl-4-phenylpyridinium (MPP+) (cell model of PD). Both in vitro and in vivo, we found that miR-30a-5p knockdown promoted glutamate uptake and increased GLT-1 expression by hindering GLT-1 ubiquitination and subsequent degradation in a PKCα-dependent manner. Therefore, we conclude that miR-30a-5p represents a potential therapeutic target for the treatment of PD.
Collapse
Affiliation(s)
- Xingjun Meng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianping Zhong
- Department of Neurology, Shunde Hospital of Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Chong Zeng
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaojuan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shaogang Qu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong 528300, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
47
|
Qin S, Tang H, Li W, Gong Y, Li S, Huang J, Fang Y, Yuan W, Liu Y, Wang S, Guo Y, Guo Y, Xu Z. AMPK and its Activator Berberine in the Treatment of Neurodegenerative Diseases. Curr Pharm Des 2021; 26:5054-5066. [PMID: 32445451 DOI: 10.2174/1381612826666200523172334] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
Neurodegenerative disorders are heterogeneous diseases associated with either acute or progressive neurodegeneration, causing the loss of neurons and axons in the central nervous system (CNS), showing high morbidity and mortality, and there are only a few effective therapies. Here, we summarized that the energy sensor adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), and its agonist berberine can combat the common underlying pathological events of neurodegeneration, including oxidative stress, neuroinflammation, mitochondrial disorder, glutamate excitotoxicity, apoptosis, autophagy disorder, and disruption of neurovascular units. The abovementioned effects of berberine may primarily depend on activating AMPK and its downstream targets, such as the mammalian target of rapamycin (mTOR), sirtuin1 (SIRT1), nuclear factor erythroid-2 related factor-2 (Nrf2), nuclear factor-κB (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), nicotinamide adenine dinucleotide (NAD+), and p38 mitogen-activated protein kinase (p38 MAPK). It is hoped that this review will provide a strong basis for further scientific exploration and development of berberine's therapeutic potential against neurodegeneration.
Collapse
Affiliation(s)
- Siru Qin
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huiling Tang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Li
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yinan Gong
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shanshan Li
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jin Huang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuxin Fang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjuan Yuan
- The First people’s hospital of Lanzhou city, Gansu, China
| | - Yangyang Liu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shenjun Wang
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongming Guo
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Guo
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhifang Xu
- Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China,Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
48
|
Dolatshahi M, Ranjbar Hameghavandi MH, Sabahi M, Rostamkhani S. Nuclear factor-kappa B (NF-κB) in pathophysiology of Parkinson disease: Diverse patterns and mechanisms contributing to neurodegeneration. Eur J Neurosci 2021; 54:4101-4123. [PMID: 33884689 DOI: 10.1111/ejn.15242] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD), the most common movement disorder, comprises several pathophysiologic mechanisms including misfolded alpha-synuclein aggregation, inflammation, mitochondrial dysfunction, and synaptic loss. Nuclear Factor-Kappa B (NF-κB), as a key regulator of a myriad of cellular reactions, is shown to be involved in such mechanisms associated with PD, and the changes in NF-κB expression is implicated in PD. Alpha-synuclein accumulation, the characteristic feature of PD pathology, is known to trigger NF-κB activation in neurons, thereby propagating apoptosis through several mechanisms. Furthermore, misfolded alpha-synuclein released from degenerated neurons, activates several signaling pathways in glial cells which culminate in activation of NF-κB and production of pro-inflammatory cytokines, thereby aggravating neurodegenerative processes. On the other hand, NF-κB activation, acting as a double-edged sword, can be necessary for survival of neurons. For instance, NF-κB activation is necessary for competent mitochondrial function and deficiency in c-Rel, one of the NF-κB proteins, is known to propagate DA neuron loss via several mechanisms. Despite the dual role of NF-κB in PD, several agents by selectively modifying the mechanisms and pathways associated with NF-κB, can be effective in attenuating DA neuron loss and PD, as reviewed in this paper.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mohammadmahdi Sabahi
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sabra Rostamkhani
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Lv N, Ma K, Li R, Liang P, Liang P, Gao X. Sublethal and lethal effects of the imidacloprid on the metabolic characteristics based on high-throughput non-targeted metabolomics in Aphis gossypii Glover. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 212:111969. [PMID: 33561773 DOI: 10.1016/j.ecoenv.2021.111969] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
Sublethal effect considered as an emerging factor to assess the environmental risk of insecticides, which can impact the insects on both physiology and behavior. Lethal exposure can be causing near immediate mortality. Pests are inevitably exposed to sublethal and lethal dose in the agroecosystem following application of pesticides. Insecticides, widely used for the control of insect pests, are irreplaceable in insect pest management. The effects of imidacloprid by the method of high-throughput non-targeted metabolomics was investigated in Aphis gossypii Glover exposed to LC10 and LC90 doses of the imidacloprid, and the control group was treated with the same condition without imidacloprid. Pairwise comparisons showed that 111 metabolites changed significantly, 60 in the LC10 group, and 66 in the LC90 group compared to the control group, while only 16 changes in the LC10 were same with that in LC90 group. Among the changed metabolites, a total of 16 metabolites were identified as potential biomarkers, which represented the most influential pathways including glycolysis and gluconeogenesis, alanine, aspartate, and glutamate metabolism, ascorbate and aldarate metabolism, glutathione metabolism, phenylalanine metabolism, tyrosine metabolism, caffeine metabolism and parkinson's disease (PD), which could account for the sublethal and lethal effects on A. gossypii. These modified metabolic pathways demonstrated that high energy consumption, excitotoxicity and oxidative stress (OS) were appeared in both LC10 and LC90 groups, while PD was detected only in the LC90 group. The results of non-targeted metabolomics revealed the effects of neonicotinoid pesticide exposure on A. gossypii successfully, and provided a deep insight into the influenced physiology by the stress of neonicotinoid pesticide in the insect.
Collapse
Affiliation(s)
- Nannan Lv
- Department of Entomology, China Agricultural University, Beijing 100193, China
| | - Kangsheng Ma
- Department of Entomology, China Agricultural University, Beijing 100193, China
| | - Ren Li
- Department of Entomology, China Agricultural University, Beijing 100193, China
| | - Pingzhuo Liang
- Department of Entomology, China Agricultural University, Beijing 100193, China
| | - Pei Liang
- Department of Entomology, China Agricultural University, Beijing 100193, China
| | - Xiwu Gao
- Department of Entomology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
50
|
Nakajima S, Saeki N, Tamano H, Nishio R, Katahira M, Takeuchi A, Takeda A. Age-related vulnerability to nigral dopaminergic degeneration in rats via Zn 2+-permeable GluR2-lacking AMPA receptor activation. Neurotoxicology 2021; 83:69-76. [PMID: 33400970 DOI: 10.1016/j.neuro.2020.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022]
Abstract
On the basis of the evidence that extracellular Zn2+ influx induced with AMPA causes Parkinson's syndrome in rats that apomorphine-induced movement disorder emerges, here we used a low dose of AMPA, which does not increase intracellular Zn2+ level in the substantia nigra pars compacta (SNpc) of young adult rats, and tested whether intracellular Zn2+ dysregulation induced with AMPA is accelerated in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome. When AMPA (1 mM) was injected at the rate of 0.05 μl/min for 20 min into the SNpc, intracellular Zn2+ level was increased in the SNpc of aged rats followed by increase in turning behavior in response to apomorphine and nigral dopaminergic degeneration. In contrast, young adult rats do not show movement disorder and nigral dopaminergic degeneration, in addition to no increase in intracellular Zn2+. In aged rats, movement disorder and nigral dopaminergic degeneration were rescued by co-injection of either extracellular (CaEDTA) or intracellular (ZnAF-2DA) Zn2+ chelators. 1-Naphthyl acetyl spermine (NASPM), a selective blocker of Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptors blocked increase in intracellular Zn2+ in the SNpc of aged rats followed by rescuing nigral dopaminergic degeneration. The present study indicates that intracellular Zn2+ dysregulation is accelerated by Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptor activation in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome.
Collapse
Affiliation(s)
- Satoko Nakajima
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Nana Saeki
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Ryusuke Nishio
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Misa Katahira
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Azusa Takeuchi
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| |
Collapse
|