1
|
de Gois AM, Bispo JMM, Santos ER, Souza MF, Melo JEC, Mendonça MS, Almeida-Souza TH, Camargo EA, Medeiros KAAL, Leal PC, Santos HF, Lins LCRF, Ribeiro AM, Silva RH, Santos JR. β-2 agonist and antagonist adrenoceptors induce neuroprotection in a progressive model of parkinsonism. Neuropharmacology 2025; 271:110386. [PMID: 40023440 DOI: 10.1016/j.neuropharm.2025.110386] [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: 09/25/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive dopaminergic dysfunction in the nigrostriatal pathway, as well as alterations in other monoamines systems. Research indicates that the use of β-adrenergic agonist and antagonists influences the risk of PD. This study evaluated the effects of salbutamol and propranolol on motor and neurochemical parameters in a progressive model of parkinsonism induced by reserpine (RES). Male Wistar rats were chronically treated with 12 subcutaneous injections of RES (0,1 mg/kg) given every other day for 24 days. From the 16th day onwards, the animals were daily treated with salbutamol (5 mg/kg) or propranolol (20 mg/kg) intraperitoneally for 8 days. Salbutamol reduced the motor deficit caused by RES in the catalepsy test, while propranolol reduced the damages caused by RES in the vacuous chewing movements evaluation. In immunohistochemical analysis both salbutamol and propranolol prevented dopaminergic damage in the substantia nigra pars compacta (SNpc), ventral tegmental area (VTA), striatum and noradrenergic damage in locus coeruleus (LC). In addition, salbutamol and propranolol prevented the increase in α-synuclein immunoreactivity caused by RES in the substantia nigra pars reticulata (SNr), striatum, prefrontal cortex (mPFC) and hippocampus. These data show that salbutamol and propranolol promote neuroprotective effects against reserpine-induced parkinsonism. However, further studies are needed to understand the mechanisms involved in β-adrenoceptors role in PD development.
Collapse
Affiliation(s)
- Auderlan M de Gois
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil.
| | - José M M Bispo
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Edson R Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Marina F Souza
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - João E C Melo
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | - Mylaine S Mendonça
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| | | | - Enilton A Camargo
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Katty A A L Medeiros
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Nursing Department, Federal University of Sergipe, Lagarto, SE, Brazil
| | - Pollyana C Leal
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Medicine, Federal University of Sergipe, Lagarto, SE, Brazil
| | - Heitor F Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Lívia C R F Lins
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Regina H Silva
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - José R Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Biosciences, Federal University of Sergipe, Itabaiana, SE, Brazil
| |
Collapse
|
2
|
Jin M, Wei Z, Ramalingam N, Xiao M, Xu A, Yu X, Song Q, Liu W, Zhao J, Zhang D, Selkoe DJ, Li S. Activation of β 2-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28:4877-4888. [PMID: 37365243 DOI: 10.1038/s41380-023-02145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of β2-adrenergic receptor (β2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid β-protein (oAβ). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aβ-species. We found that pharmacological activation of β2-AR, but not β1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAβ-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in β2-AR knockout mice, suggesting that activating β2-AR prevents oAβ-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of β-ARs each decreased HDAC2, whereas Aβ oligomers increased HDAC2 levels in the hippocampus. Further, oAβ-induced inflammatory effects and neurite degeneration were prevented by either β2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of β2-AR is a novel potential therapeutic strategy to mitigate oAβ-mediated features of AD.
Collapse
Affiliation(s)
- Ming Jin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Meng Xiao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China
| | - Anqi Xu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Xiaohan Yu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Qingyang Song
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zhao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Henan Key Laboratory of Neurorestoratology, Xinxiang, Henan, 453100, China
| | - Dainan Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Pilipović I, Stojić-Vukanić Z, Leposavić G. Adrenoceptors as potential target for add-on immunomodulatory therapy in multiple sclerosis. Pharmacol Ther 2023; 243:108358. [PMID: 36804434 DOI: 10.1016/j.pharmthera.2023.108358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
This review summarizes recent findings related to the role of the sympathetic nervous system (SNS) in pathogenesis of multiple sclerosis (MS) and its commonly used experimental model - experimental autoimmune encephalomyelitis (EAE). They indicate that noradrenaline, the key end-point mediator of the SNS, acting through β-adrenoceptor, has a contributory role in the early stages of MS/EAE development. This stage is characterized by the SNS hyperactivity (increased release of noradrenaline) reflecting the net effect of different factors, such as the disease-associated inflammation, stress, vitamin D hypovitaminosis, Epstein-Barr virus infection and dysbiosis. Thus, the administration of propranolol, a non-selective β-adrenoceptor blocker, readily crossing the blood-brain barrier, to experimental rats before the autoimmune challenge and in the early (preclinical/prodromal) phase of the disease mitigates EAE severity. This phenomenon has been ascribed to the alleviation of neuroinflammation (due to attenuation of primarily microglial activation/proinflammatory functions) and the diminution of the magnitude of the primary CD4+ T-cell autoimmune response (the effect associated with impaired autoantigen uptake by antigen presenting cells and their migration into draining lymph nodes). The former is partly related to breaking of the catecholamine-dependent self-amplifying microglial feed-forward loop and the positive feedback loop between microglia and the SNS, leading to down-regulation of the SNS hyperactivity and its enhancing influence on microglial activation/proinflammatory functions and the magnitude of autoimmune response. The effects of propranolol are shown to be more prominent in male EAE animals, the phenomenon important as males (like men) are likely to develop clinically more severe disease. Thus, these findings could serve as a firm scientific background for formulation of a new sex-specific immune-intervention strategy for the early phases of MS (characterized by the SNS hyperactivity) exploiting anti-(neuro)inflammatory and immunomodulatory properties of propranolol and other relatively cheap and safe adrenergic drugs with similar therapeutic profile.
Collapse
Affiliation(s)
- Ivan Pilipović
- Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- University of Belgrade-Faculty of Pharmacy, Department of Microbiology and Immunology, Belgrade, Serbia
| | - Gordana Leposavić
- University of Belgrade-Faculty of Pharmacy, Department of Pathobiology, Belgrade, Serbia.
| |
Collapse
|
4
|
Li S. The β-adrenergic hypothesis of synaptic and microglial impairment in Alzheimer's disease. J Neurochem 2023; 165:289-302. [PMID: 36799441 DOI: 10.1111/jnc.15782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease originating partly from amyloid β protein-induced synaptic failure. As damaging of noradrenergic neurons in the locus coeruleus (LC) occurs at the prodromal stage of AD, activation of adrenergic receptors could serve as the first line of defense against the onset of the disease. Activation of β2 -ARs strengthens long-term potentiation (LTP) and synaptic activity, thus improving learning and memory. Physical stimulation of animals exposed to an enriched environment (EE) leads to the activation of β2 -ARs and prevents synaptic dysfunction. EE also suppresses neuroinflammation, suggesting that β2 -AR agonists may play a neuroprotective role. The β2 -AR agonists used for respiratory diseases have been shown to have an anti-inflammatory effect. Epidemiological studies further support the beneficial effects of β2 -AR agonists on several neurodegenerative diseases. Thus, I propose that β2 -AR agonists may provide therapeutic value in combination with novel treatments for AD.
Collapse
Affiliation(s)
- Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Wu L, Tai Y, Hu S, Zhang M, Wang R, Zhou W, Tao J, Han Y, Wang Q, Wei W. Bidirectional Role of β2-Adrenergic Receptor in Autoimmune Diseases. Front Pharmacol 2018; 9:1313. [PMID: 30538630 PMCID: PMC6277539 DOI: 10.3389/fphar.2018.01313] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
Disorder of the sympathetic nervous system (SNS) is closely related to the pathogenesis of various autoimmune diseases (ADs). Catecholamine triggered beta2-adrenergic receptor (β2-AR) signaling is important in creating a bidirectional response in the progression of ADs due to factors including diverse expression patterns, single nucleotide polymorphisms (SNPs), biased signals, and desensitization of β2-AR, as well as different subtypes of Gα binding to β2-AR. In this review, we summarize the actions of β2-AR signaling in regulating the functions of immunocytes and in the pathogenesis of ADs, and the application of β2-AR agonists or antagonists in treating major types of ADs is also discussed. We suggest that restoring the immune balance via a soft regulation of the expression or activation of β2-AR is one of the promising therapeutic strategies for systematic ADs.
Collapse
Affiliation(s)
- Li Wu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Tai
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Shanshan Hu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Mei Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Rui Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Weijie Zhou
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Juan Tao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| | - Qingtong Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Mahaman YAR, Huang F, Kessete Afewerky H, Maibouge TMS, Ghose B, Wang X. Involvement of calpain in the neuropathogenesis of Alzheimer's disease. Med Res Rev 2018; 39:608-630. [PMID: 30260518 PMCID: PMC6585958 DOI: 10.1002/med.21534] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/11/2018] [Accepted: 07/29/2018] [Indexed: 01/02/2023]
Abstract
Alzheimer’s disease (AD) is the most common (60% to 80%) age‐related disease associated with dementia and is characterized by a deterioration of behavioral and cognitive capacities leading to death in few years after diagnosis, mainly due to complications from chronic illness. The characteristic hallmarks of the disease are extracellular senile plaques (SPs) and intracellular neurofibrillary tangles (NFTs) with neuropil threads, which are a direct result of amyloid precursor protein (APP) processing to Aβ, and τ hyperphosphorylation. However, many indirect underlying processes play a role in this event. One of these underlying mechanisms leading to these histological hallmarks is the uncontrolled hyperactivation of a family of cysteine proteases called calpains. Under normal physiological condition calpains participate in many processes of cells’ life and their activation is tightly controlled. However, with an increase in age, increased oxidative stress and other excitotoxicity assaults, this regulatory system becomes impaired and result in increased activation of these proteases involving them in the pathogenesis of various diseases including neurodegeneration like AD. Reviewed here is a pool of data on the implication of calpains in the pathogenesis of AD, the underlying molecular mechanism, and the potential of targeting these enzymes for AD therapeutics.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Henok Kessete Afewerky
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tanko Mahamane Salissou Maibouge
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bishwajit Ghose
- Department of Social Medicine and Health Management, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Division of Neurodegenerative Disorders, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
7
|
Smith AW, Ray SK, Das A, Nozaki K, Rohrer B, Banik NL. Calpain inhibition as a possible new therapeutic target in multiple sclerosis. AIMS MOLECULAR SCIENCE 2017; 4:446-462. [PMID: 40181912 PMCID: PMC11967729 DOI: 10.3934/molsci.2017.4.446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Multiple sclerosis (MS), the most common chronic autoimmune inflammatory disease of the central nervous system (CNS), is characterized by demyelination and neurodegeneration. In particular, neurodegeneration is a major factor in disease progression with neuronal death and irreversible axonal damage leading to disability. MS is manageable with current therapies that are directed towards immunomodulation but there are no available therapies for neuroprotection. The complex pathophysiology and heterogeneity of MS indicate that therapeutic agents should be directed to both the inflammatory and neurodegenerative arms of the disease. Activity of the Ca2+ activated protease calpain has been previously implicated in progression of MS and its primary animal model, experimental autoimmune encephalomyelitis (EAE). The effects of calpain inhibitors in EAE involve downregulation of Th1/Th17 inflammatory responses and promotion of regulatory T cells, overall leading to decreased inflammatory cell infiltration in CNS tissues. Furthermore, analysis of brains, spinal cords and optic nerves from EAE animals revealed decreases in axon degeneration, motor neuron and retinal ganglion cell death. This resulted in improved severity of paralysis and preservation of visual function. Taken together, the studies presented in this brief review suggest that use of calpain inhibitors in combination with an immunomodulatory agent may be a potential therapeutic strategy for MS and optic neuritis.
Collapse
Affiliation(s)
- Amena W. Smith
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, SC, USA
| | - Arabinda Das
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Kenkichi Nozaki
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Naren L. Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|